You are on page 1of 14

European Journal of Pharmacology 775 (2016) 1–14

Contents lists available at ScienceDirect

European Journal of Pharmacology


journal homepage: www.elsevier.com/locate/ejphar

Review

Cannabinoid pharmacology in cancer research: A new hope


for cancer patients?
Farideh A. Javid a,n, Roger M. Phillips a, S. Afshinjavid b, Roberta Verde c, Alessia Ligresti c
a
Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield HD1 3DH, UK
b
Faculty of Science and Engineering, University of Chester, Pool lane, Chester CH2 4NU, UK
c
Endocannabinoid Research Group, Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, NA, Italy

art ic l e i nf o a b s t r a c t

Article history: Cannabinoids have been used for many centuries to ease pain and in the past decade, the endocannabinoid
Received 4 June 2015 system has been implicated in a number of pathophysiological conditions, such as mood and anxiety dis-
Received in revised form orders, movement disorders such as Parkinson's and Huntington's disease, neuropathic pain, multiple
5 January 2016
sclerosis, spinal cord injury, atherosclerosis, myocardial infarction, stroke, hypertension, glaucoma, obesity,
Accepted 3 February 2016
and osteoporosis. Several studies have demonstrated that cannabinoids also have anti-cancer activity and as
Available online 5 February 2016
cannabinoids are usually well tolerated and do not produce the typical toxic effects of conventional che-
Keywords: motherapies, there is considerable merit in the development of cannabinoids as potential anticancer
Cannabinoids therapies. Whilst the presence of psychoactive effects of cannabinoids could prevent any progress in this
Cancer field, recent studies have shown the value of the non-psychoactive components of cannabinoids in activating
Cannabinoid receptors
apoptotic pathways, inducing anti-proliferative and anti-angiogenic effects. The aforementioned effects are
suggested to be through pathways such as ERK, Akt, mitogen-activated protein kinase (MAPK) pathways,
phosphoinositide 3-kinase (PI3K) pathways and hypoxia inducible factor 1 (HIF1), all of which are important
contributors to the hallmarks of cancer. Many important questions still remain unanswered or are poorly
addressed thus necessitating further research at basic pre-clinical and clinical levels. In this review, we ad-
dress these issues with a view to identifying the key challenges that future research needs to address.
& 2016 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Anti-tumour effects of cannabinoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3

Abbreviations: ABCC1, ATP-binding cassette (ABC) transporter; AC, adenylyl cyclase; AEA, anandamide; AKt, protein kinase B; AM251, a CB1 receptor antagonist; AM630, a
CB2 receptor antagonist; AMPK, 5′-adenosine monophosphate-activated protein kinase; ATF-4, activating transcription factor-4; AR, androgen receptors; CBD, cannabidiol;
CB1IR, CB1 receptor immunoreactivity; Cdk, cyclin-dependent kinase; Chk 1, cell cycle checkpoint; COX2, cyclooxygenase-2; CXCR4, chemokine receptor 4; CXCL12, a
chemokine protein encoded by the CXCL12 gene; Δ9-THC, Δ9-tetrahydrocannabinol; EGFR, epidermal growth factor receptor; ER, estrogen; ERK, extracellular signal-
regulated kinase; FAAH, fatty acid amide hydrolase; FAK, focal adhesion kinase; GBM, glioblastoma multiform; Gi/o, a subunit of G protein; GTPγS, guanosine 5′-O-[gamma-
thio] triphosphate; HER2, human epidermal growth factor receptor 2; HIF-α, hypoxia-inducible factor; HU-210, highly potent cannabinoid receptor agonist, 96aR0-trans-3–
91, 1-Dimethylheptyl)-6a, 7, 10, 10a-tetrahydro-1-hydroxy-6, 6-dimethyl-6H-dibenzo [b,d]pyran-9-methanol; ICAM, intracellular adhesion molecule; JWH-015, a selective
CB2 agonist, 2-Methyl-1-propyl-1H-indol-3-yl)-1-naphthalenylmethanone; JWH-133, a potent selective CB2 agonist, (6aR, 10aR)-3-(1,1-dimethylbutyl)-6a,7,10,10a-Tetra-
hydro-6,6,9-trimethyl-6H-dibenzo [b,d]pyran MAPK, mitogen activated protein kinase; LAK, lymphokine-activated killer; LPI, lysophosphatidylinositol; MAGL, monoglycerol
lipase; MMP-2, matrix metallopeptidase 2; MMP-9, matrix metallopeptidase 9; MRP1, Multidrug resistance-related protein 1; mTOR, mammalian target of rapamycin; NF-
κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NGF, nerve growth factor; PD98059, p42/44 inhibitor, 2-(2-Amino-3-methoxyphenyl)-4/h-1-benzopyran-
4-one; PGE2, prostaglandine E-2; PI3K, phosphoinositide 3-kinase; PKA, protein kinase A; PCNA, proliferating cell nuclear antigen; PPARs, peroxisome proliferator-activated
receptors; PR, progesterone; PRLr, prolactin receptor; PSA, prostate specific antigen; PyMT, polyoma middle T oncoprotein; RAF-1, a proto-oncogene, serine/threonine
kinase; ROS, reactive oxygen species; RXRα, retinoid X receptor; SB203580, a p38/MAPK inhibitor, 4-[5-(4-Fluorophenyl)-2-[4-(methylsulphonyl)phenyl]-1H-imidazol-4-yl]
pyridine; SC58236, COX-2-specific inhibitor, 4-[5-(4-chlorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]benzenesulfonamide; siRNA, small interfering RNA; SR141716, Ri-
monabant, a selective CB1 receptor antagonist or an inverse agonist; Src gene, a family of proto-oncogenic tyrosine kinases; Th1, a type of T helper cells; Th2, a type of T
helper cells; TIMP-1, tissue inhibitor of matrix metalloproteinases-1; TrK A, tropomyosin receptor kinase A; TRPM8, transient receptor potential channels of melastatin-type
8; TRPVA1, transient receptor potential A1; TRPV1, transient receptor potential vanilloid 1; TRB3, tribbles homologue that inhibits Akt/PKB activation; 2-AG, 2-arachidonoyl
glycerol; VEGF, vascular endothelial growth factor; WIN 55,212–2, a CB1 and CB2 receptor agonist [(R0-( þ )-[2,3-Dihydro-5-methyl-3-(4-morpholinylmethyl) pyrrolo[1,2,3-
de]-1,4-benzoxazin-6-yl]-1-naphtalenylmethanoneesylate]
n
Correspondence to: Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK.
E-mail address: fajavid@hud.ac.uk (F.A. Javid).

http://dx.doi.org/10.1016/j.ejphar.2016.02.010
0014-2999/& 2016 Elsevier B.V. All rights reserved.
2 F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14

3. Cannabinoids and breast cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4


4. Cannabinoids and brain cancer. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5. Cannabinoid and lung cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
6. Cannabinoids and intestinal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
7. Cannabinoids and reproductive system cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
8. Conclusion and future direction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Conflict of interests/acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10

1. Introduction liver disorder, pain and atherosclerosis (Buckley, 2008).


Both CB1 and CB2 receptors are metabotropic and belong to the
It is known that cannabinoids, the active components of Can- G-protein coupled receptor family (Howlett et al., 2002). Activa-
nabis sativa, act by mimicking the endogenous substances (the tion of CB1 and CB2 receptors stimulates cellular signalling via
endocannabinoids anandamide and 2-arachidonoylglycerol (2- alpha subunit of G protein (Gi/o), leading to inhibition of adenylate
AG)) by activating specific cell-surface cannabinoid receptors cyclase and the subsequent activation of many other pathways
(Devane et al., 1992). Currently, the cannabinoid receptor ligands such as mitogen-activated protein kinase (MAPK) pathways,
are generally divided into three main categories known as phy- phosphoinositide 3-kinase (PI3K) pathways, modulation of ion
tocannabinoids, endogenous cannabinoids and synthetic canna- channels (through CB1 receptors), protein kinase B (Akt), ceramide
binoids (Fig. 1). After the clarification of the chemical structure of signalling pathways in tumour cells and modulation of cycloox-
(-)-Δ9-tetrahydrocannabinol (Δ9-THC) which is the primary psy- ygenase-2 (COX-2) signalling pathway (Demuth and Molleman,
choactive component of the cannabis plant (Gaonia and Mechou- 2006; Galve-Roperh et al., 2000; Glass and Northup, 1999; Guz-
lam, 1964a, 1964b), other chemically related terpenophenolic man et al., 2001; Qamri et al., 2009).
compounds were identified in Cannabis sativa, including canna- There is also pharmacological evidence that non-CB1 and non-CB2
bichromene (CBC) (Gaoni and Mechoulam, 1966) and cannabigerol receptors mediate the actions of cannabinoids located in the brain
(CBG) (Gaoni and Mechoulam, 1964c). Although the pharmacology (Breivogel et al., 2001; Di Marzo et al., 2000). The hypothesis that
of most of the cannabinoids is unknown, Δ9-THC is the most putative CB3 or non-CB1/CB2 receptor exist is supported by the fact that
widely studied owing to its high potency and abundance in can- some of the anandamide (AEA)-mediated effects were neither in-
nabis (Pertwee et al., 2010). Among the herbal cannabinoids, other hibited by selective CB antagonists nor fully abolished in knockout mice
relevant plant-derived cannabinoids include Δ8-THC, which is al- lacking CB1 receptors (De Petrocellis and Di Marzo, 2010). Recent ad-
most as active as Δ9-THC but less abundant and cannabinol (CBN), vances suggest, at least for AEA, that the transient receptor potential
which is produced in large amounts but is a weak cannabomimetic vanilloid 1 receptor (TRPV1) channel may be considered as the “third”
agent. Cannabidiol (CBD), CBG and CBC are devoid of psychoactive receptor involved in endocannabinoid signalling (Di Marzo et al., 2001;
potential. The chemical structures of some cannabinoids are Ross, 2003). For example, it has been shown that the endocannabinoids
shown in Fig. 1. exert their apoptotic effect by binding to TRPV1, a non-selective cation
So far, two cannabinoid-specific receptors CB1 and CB2 have channel targeted by capsaicin, the active component of hot chilli pep-
been cloned and characterized from mammalian tissues (Howlett pers (Smart and Jerman, 2000). However, the precise role of this re-
et al., 2002). Mouse CB1 receptor and CB2 receptor share 66% ceptor in cannabinoid signalling is still unclear and this uncertainty
overall homology and 78% in the transmembrane region (Shire extends into the cancer field where its potential role in cancer biology
et al., 1996). Human CB1 receptor and CB2 receptor share an overall (proliferation and migration of cancer cells) and cancer pharmacology
homology of 44%, and 68% in the transmembrane region respec- (resistance to chemotherapeutic agents) needs further investigation
tively (Munro et al., 1993). Homology (96%) has been reported (Lehen'kyi and Prevarskaya, 2011; Liberati et al., 2013). Evidence also
between human and mouse CB1 receptor (Chakrabarti et al., 1995), exists supporting a role for peroxisome proliferator-activated receptors
whilst human and mouse CB2 receptors share 82% homology (PPARs) in the actions of cannabinoids (Sun and Bennett, 2007). More
(Shire et al., 1996). Many central and peripheral effects have been recent studies have provided evidence for the interaction of cannabi-
associated with the activation of CB1-receptors (Matsuda et al., noids with the orphan receptors such as G protein receptor 55 (GPR55)
1990; Munro et al., 1993; Pertwee, 2006; Pertwee et al., 2010). The (Andradas et al., 2011; Pineiro et al., 2011). Thus in addition to CB1 and
CB2 receptor, originally thought of as being exclusively present in CB2 receptors other targets might be involved in mediating an effect to
the immune system, is highly expressed in B and T lymphocytes, cannabinoids and endocannabinoids.
macrophages and in tissues such as the spleen, tonsils and lymph The potential of cannabinoids to alleviate pain has been re-
nodes (Herkenham et al., 1991; Howlett et al., 2002; Porter and cognised for many centuries. The antinociceptive actions are
Felder, 2001; Pertwee et al., 2010). Recently CB2 receptors have mediated via both the CB1 and CB2 receptors (Pacher et al., 2006).
been shown to be also located in the brain stem (Van Sickle et al., This does not negate a role for other receptors such as TRPV1,
2005). Further studies using CB1 knockout mice demonstrated that transient receptor potential cation channel A1(TRPA1), orphan
CB1 receptors are involved in a variety of different behavioural GPCR (i.e. GPR55) or PPAR-γ (Maione et al., 2006, 2013; Perez-
disorders such as depression, anxiety, feeding and cognition as Gomez et al., 2013; Moreno et al., 2014). For a long time, the de-
well as pain at the peripheral, spinal and supraspinal levels (Val- velopment of cannabinoids as anticancer agents has been re-
verde et al., 2005). Such studies using CB1 knockout mice also stricted to two therapeutic avenues (antiemetic and analgesic).
revealed the interactions between different systems such as They have therefore been evaluated in terms of palliative care as
opioids, gamma aminobutyric acid (GABA) and cholecystokinin cannabinoids can play an important role in the relief of pain,
(CCK) via CB1 receptors (Valverde et al., 2005). CB2 knockout mice nausea, vomiting, and stimulation of appetite in cancer patients.
have also been developed and revealed/confirmed the involve- However, the involvement of CB receptors in pain and their use in
ment of CB2 receptors in a variety of different systems such as the palliative care in cancer patients are not the focus of this re-
immune system, inflammation, apoptosis, chemotaxis, bone loss, view. In the present review, the aim is to focus on the anti-tumour
F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14 3

Phytocann
nabinoids:

Cannaabidiol (CBD
D) Delta-9- Tetrahydrocaannabinol (Δ9-THC)

Synthetic cannabinoids:

HU 210 WIN 552212-2

Endocann
nabinoids:

Ananndamide (A
AEA) 2-AG
Fig. 1. Chemical structures of some cannabinoids.

effects of cannabinoids, identify potential mechanisms by which to report the anti-proliferative properties of cannabis compounds
cannabinoids induce anti-tumour effects and discuss the potential (Munson et al., 1975). They showed that Δ9-THC inhibits lung-
and challenges for the future development of this class of adenocarcinoma cell growth in vitro and after oral administration
compound. in mice. It was not until the late 1990s however that further stu-
dies on the anti-cancer effects of cannabinoids were carried out.
Several plant-derived (for example, Δ9-THC and CBD), synthetic
2. Anti-tumour effects of cannabinoids (for example, WIN-55, 212–2 and HU-210) and endogenous can-
nabinoids (for example, AEA and 2-AG) are now known to exert
Whilst cannabinoids exert palliative effects in cancer patients anti-proliferative actions on a wide range of tumour cells in vitro
by preventing nausea, vomiting and pain and by stimulating ap- (Guzman et al., 2002). The involvement of CB1 and/or CB2 re-
petite, they have also been shown to inhibit the growth of tumour ceptors in the anti-tumour effects of cannabinoids has been shown
cells in culture and animal models by modulating key cell-sig- by various biochemical and pharmacological approaches, in par-
nalling pathways. In 1975, Munson and collaborators were the first ticular by determining cannabinoid-receptor expression and by
4 F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14

using selective cannabinoid-receptor agonists and antagonists proliferation of PC-3 cells (Melck et al., 2000) through interaction
(Guzmán, 2003; Sarfaraz et al., 2008; Pisanti et al., 2013 ;Velasco with CB1 receptors and synthetic endocannabinoid-vanilloid hy-
et al., 2012). Such studies have shown that cannabinoids can brids via stimulation of TRPV1 channels. However, other studies
prevent proliferation, metastasis, angiogenesis and exert pro- demonstrate that cannabinoid-induced anti-tumour activity was
apoptotic effects in a variety of cancer cell types such as lung, only marginally dependent upon the interaction between canna-
breast, prostate, skin, intestine, glioma, lymphoma, pancreas and binoid and TRPV1 receptors (Massi et al., 2004; Torres et al., 2011;
uterus (Blazquez et al., 2006; Carracedo et al., 2006b; Casanova Vaccani et al., 2005). On the other hand, cannabinoids have also
et al., 2003; Cianchi et al., 2008; Galve-Roperh et al., 2000; Guz- been shown to actively induce apoptosis in these cells via a CB1
mán, 2003; Pacher et al., 2006; Sánchez et al., 2001a, 2001b). Si- receptor-independent mechanism as the Δ9-THC-induced apop-
lencing CB2 receptors with specific small interfering RNA (siRNA) tosis was not reversed by the CB1 receptor antagonist, SR141716
in prostate cancer cells (PC-3) revealed the involvement of CB2 (Ruiz et al., 1999).
receptors in the growth inhibition of prostate cancer cells (Olea- The effects of cannabinoids in modulating cell cycle and sig-
Herrero et al., 2009) via stimulation of autophagy. Cannabinoid- nalling pathways are diverse (Fig. 2) and may depend on the type
induced AMPK activation of autophagy was also confirmed in vivo of tumour cells. Whilst there is evidence showing that some
(Vara et al., 2011). The over expression of cannabinoid receptors cannabinoids induce anti-proliferative effect on tumour cells in
and elevated endocannabinoid levels have also been reported in a vitro (Hart et al., 2004; White et al., 1976; Velasco et al., 2015),
variety of different cancer types such as prostate, skin, hepato- there is evidence to suggest that the effects may depend upon the
cellular carcinoma, colon, endometrial sarcoma, glioblastoma disease context with differential effects seen in different tumour
multiforme (GBM), meningioma and pituitary adenoma (see Ta- types. The subsequent sections focus specifically on the evidence
ble 1) (Blazquez et al., 2006; Xu et al., 2006; Pisanti et al., 2013). of cannabinoid induced anti-tumour effects in specific cancer
CB1 receptors are also up-regulated in Hodgkin lymphoma cells types with the aim of exploring the mechanism of action of non-
and also in chemically induced hepatocarcinoma (Benz et al., 2013; psychoactive components of cannabinoids in a disease specific
Mukhopadhyay et al., 2015). Also the expression of CB1 and CB2 context.
receptors was found to increase in mantel cell lymphoma and fatty
acid amide hydrolase (FAAH) expression was reduced when
compared to non-malignant B-cells (Islam et al., 2003; Ek et al., 3. Cannabinoids and breast cancer
2002; Wasik et al., 2014). It has also been shown in both the
mouse model of metastatic melanoma and in humans that the The first report on the antineoplastic property of cannabinoids
circulating endocannabinoid levels have been associated with an in breast cancer are in the late 1990s, when it was shown that pre-
increase in disease progression (Sailler et al., 2014). Indeed many treatment with the endocannabinoid anandamide inhibited pro-
reports have shown that an increase in the level of en- lactine- and nerve growth factor-induced proliferation of two
docannabinoids and their receptors correlates with tumour ag- hormone-sensitive, estrogen and progesterone (ER þ /PR þ ) breast
gressiveness (Malfitano et al., 2011) and that cannabinoids can cancer cell lines (EFM-19 and MCF-7 cell lines). In this case,
inhibit the growth of xenograft tumours (Blazquez et al., 2006; treatment reduced the levels of prolactin receptor (PRLr) and
Carracedo et al., 2006b; Sánchez et al., 2001a). nerve growth factor receptors via CB1 receptor activation. Thus,
In neoplastic cells cannabinoids have also been shown to in- the inhibition of adenylyl cyclase activity that in turn induced
hibit angiogenesis and directly initiate apoptosis or cell cycle ar- prolonged activation/stimulation of the RAF1-MEK-ERK cascade,
rest (Blazquez et al., 2006; Carracedo et al., 2006b; Sánchez et al., leads to a down-regulation of the PRL receptor and levels of TrkA
2001a). Other studies have shown the ability of cannabinoids to NGF receptors (De Petrocellis et al., 1998; Melck et al., 2000, 1999).
affect cellular signalling pathway/s essential for cell survival and Of interest, other studies showed that in both MCF-7 cells and
growth (Bifulco et al., 2008; Kogan, 2005). Some studies have also tamoxifen-resistant MCF-7 (TAMR-MCF-7) tumour cells, anti-an-
demonstrated and suggested the involvement of autophagy in the giogenesis effects exerted by novel synthetic hexahydrocannabinol
mechanism of cannabinoid-induced cytotoxicity (Armstrong et al. analogueues was through the suppression of VEGF (Thapa et al.,
2015; Vara et al., 2011; McAllister et al., 2015). Cannabinoids have 2011). It should be noted that both MCF-7 and TAMR-MCF-7 cells
also been reported to inhibit nerve growth factor (NGF)-induced have shown a strong association between enhanced VEGF

Table 1
Tumours, which are sensitive to cannabinoid-induced growth inhibition.

Tumour type Experimental system Effect Receptor Ref.

Lung carcinoma In vivo (mouse) Decreased tumour size N.D. Munson et al. (1975)
In vitro (isolated lewis lung cells) Cell-growth
inhibition
Glioma In vivo (mouse, rat) Decreased tumour size CB1, CB2 Galve-Roperh et al. (2000), Sánchez et al. (2001a) and Ja-
In vitro (C6 glioma cells) Apoptosis cobsson et al. (2000)
Thyroid epithelioma In vivo (mouse) Decreased tumour size CB1 Bifulco et al. (2001)
In vitro (KiMol cells) Cell-cycle arrest
Lymphoma/leukaemia In vivo (mouse) Decreased tumour size CB1/CB2 McKallip et al. (2002) and Gustafsson et al. (2006)
In vitro (MCL cells; Jurkat cells; Molt-4 cells; Apoptosis
Sup-T1 cells)
Skin carcinoma In vivo (mouse) Decreased tumour size CB1, CB2 Casanova et al. (2003)
In vitro (PDV.C57 cells; HaCa4 cells) Apoptosis
Uterus carcinoma In vitro (HeLa cells) Cell-growth inhibition N.D. Mon et al. (1978), Blevins and Smith (1980)
Breast carcinoma In vitro (MCF-7 cells) Cell-cycle arrest CB1 DE Petrocellis et al. (1998), Melck et al. (2000)
Prostate carcinoma In vitro (LNCaP cells; DU-145 cells; PC3 cells) Apoptosis CB1/CB2 Mimeault et al. (2003), Melck et al. (2000), Ruiz et al. (1999),
Sarfaraz et al. (2005)
Neuroblastoma In vitro (CHP100 cells) Apoptosis Trpv1 Maccarrone et al. (2000)

N.D., not determined; TRPV1, type 1 vanilloid receptor.


F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14 5

Activation of cannabinoids receptors

N N

C C
c-AMP EGFR Activation of Ceramide

PKA phosphorylation of ERK1/2 , JNK1/2 , Akt, FAK ERK1/2 PI3K and Akt Bad protein p38 p8

FAK tyrosine phosphorylation Cyclins ATF-4, CHOP and TRB3


MAPK activation

Inactivation of tyrosine phosphatases Cdks caspases

Anti-proliferative Migration
effects Migration,
invasion & growth

Apoptosis

Fig. 2. Schematic representation showing some examples of pathways activated following the activation of cannabinoid receptors.

Agonists/endocannabinoids binding at cannabinoid receptors

N N

C C
cAMP erbB Ras loss in Cdk2 activity Cox-2 Chk1 activation and
Cdc25A proteolysis

PKA RAF1 p2/waf PGE2 dephosphorylation


through RAF1/MEK/ERK cyclin E/Cdk2 kinase of Thr14/Tyr15
cascade activation

MAPK PRL and trk level prevention of Cdk2 activation


activation

Anti-proliferative Angiogenesis Cell arrest in S phase of


effect & apoptosis the cell cycle
Anti-proliferative
effect at the S
phase

Fig. 3. Schematic representation of examples of different pathways associated with anti-proliferative effects induced by cannabinoid receptor activation in breast cancer.

production and more aggressive phenotype (Kim et al., 2008, role in hormone-sensitive breast cancer cell migration and inva-
2009). The anti-angiogenesis activity afforded by the novel syn- sion. Specifically, CB2 has recently been found to modulate breast
thetic cannabinoids were shown to be independent of CB1 and CB2 tumour growth and metastasis by inhibiting signalling of the
receptor activity and through an inhibition of NF-κB transcrip- chemokine receptor CXCR4 and its ligand CXCL12 in both in MCF7
tional activity which in turn plays an important role in VEGF overexpressing CXCR4 and NT2.5 injected immune-competent
regulation and angiogenesis (Thapa et al., 2011). Further studies syngenic FVB mice (Nasser et al., 2011). However, other studies
demonstrated that the proliferation of EVSA-T, a hormone-sensi- showed that Δ9-THC enhanced breast cancer growth and metas-
tive (ER  /PR þ ) breast cancer cell line, was also inhibited by TCH tasis although specifically in cells expressing low levels of canna-
via CB2 receptor activation. This caused the activation of the binoid receptors (i.e. mouse mammary carcinoma 4T1 cells) by
transcription factor JunD, the up-regulation of gene expression suppressing the antitumour immune response (McKallip et al.,
and subsequent translocation of protein to the nuclear compart- 2005). Such studies showed that exposure to Δ9-THC led to an
ment (Caffarel et al., 2008). Thus, it seems that cannabinoids do increase in the level of cytokines such as IL-4 and IL-10, suggesting
activate many cell-specific pathways, however not all pathways that Δ9-THC exposure may specifically suppress the cell-mediated
are simultaneously stimulated (Fig. 3). In addition to their anti- Th1 response by enhancing Th2-associated cytokines (McKallip
mitogenic property, cannabinoids have been also shown to play a et al., 2005). Among the hormone-sensitive histopathological
6 F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14

subtypes, it should be mentioned that in breast tumours that ex- stress, all contributing to induce apoptosis. The efficacy of CBD was
press the tyrosine kinase receptor HER2, cannabinoids have been also corroborated with in vivo data in athymic mice injected with
shown to be very effective. In fact, in addition to the correlation human MDA-MB-231 breast carcinoma cells (Ligresti et al., 2006).
between tumour aggressiveness and CB2 receptor expression in Indeed, it was also shown that in MDA–MB231, a human breast
breast cancer, a significant correlation between CB2 receptor and cell line, CBD induced endoplasmic reticulum stress, inhibition of
ErbB2 has been recently demonstrated (Caffarel et al., 2010). In AKT/mTOR pathway, and up-regulation of autophagy-mediated
particular, this study showed that the selective agonist JWH-133 cell death (Shrivastava et al., 2011). Recent studies indicated an
was as effective as Δ9-THC (a CB1/CB2-mixed agonist) in reducing anticancer activity induced by a quinone/cannabinoid derivative
tumour growth and progression through inhibition of the pro- through the activation of CB2 receptors and oxidative stress me-
tumourigenic kinase Akt pathway. Moreover, in vivo studies re- chanism to induce apoptosis in triple-negative breast cancer, a
ported that Δ9-THC was efficacious in reducing tumour growth, highly aggressive type of breast cancer (Morales et al., 2015). Other
tumour number, and the amount/severity of lung metastases in recent studies indicated that CBD significantly inhibits epidermal
MMTV-neu mice (Caffarel et al., 2010). growth factor (EGF)-induced proliferation and chemotaxis of
Another group of clinically important breast tumours are triple- breast cancer cells. In addition, it was shown that CBD inhibits
negative tumours. These tumours are characterized by the total EGF-induced activation of EGFR, ERK, AKT and NF-kB signalling
lack of expression in ER, PR or HER2. Numerous pieces of evidence pathways as well as MMP2 and MMP9 secretion (Elbaz et al.,
both in vitro and in vivo, demonstrate that cannabinoids (acting 2015). CBD also inhibited tumour growth and metastasis in dif-
through a plethora of different mechanisms) can be considered as ferent mouse model systems. Analysis of molecular mechanisms
promising candidates for the treatment of ER  /PR  /HER2  breast revealed that CBD significantly inhibits the recruitment of tumour-
cancer. The metabolically stable anandamide analogueue (Met-F- associated macrophages in primary tumour stroma and secondary
AEA), significantly affected adhesion and migration of both the lung metastases (Elbaz et al., 2015). Fig. 3 summarizes the im-
highly invasive human breast carcinoma cell line (MDA-MB-231) portant pathways in cannabinoid induced cytotoxicity.
and murine breast cancer cell line (TSA-E1) by reducing FAK tyr-
osine phosphorylation/activation and Src phosphorylation via CB1
receptor (Grimaldi et al., 2006). Interestingly, the same group re- 4. Cannabinoids and brain cancer
ported that pre-treatment with SR141716 could also inhibit tu-
mour growth or induce apoptosis possibly through an inhibition of Both CB1 and CB2 receptors have been identified in the CNS
ERK1/2 signalling in lipid rafts and caveolae (Sarnataro et al., (Ameri, 1999; Benito et al., 2007; Herkenham et al., 1991; Nunez
2006). Both are highly implicated in tumour growth and metas- et al., 2004; Skaper et al., 1996). High density of CB1 receptors has
tasis in breast cancer (Sloan et al., 2004; Williams et al., 2004). been reported in different areas of the brain such as in the cortex,
Later studies reported an over- expression of cannabinoid re- cerebellum and hippocampus (Herkenham et al., 1991; Hoffman
ceptors in primary human breast tumours compared with normal et al., 2010; Sullivan, 2000; Tsou et al., 1998). The CB1 receptor
breast tissues, as well as in breast cancer cell lines MDA–MB231 protein is mainly localised in astroglial cells and neurones whereas
and MDA–MB468 (Qamri et al., 2009). Such studies have shown CB2 receptors are located on microglial cells (Held-Feindt et al.,
that stimulation of both CB1 and CB2 receptor by their agonists, 2006; Stella, 2004) with possible neuroprotective activity (Cabral
WIN-55,212–2 and JWH-133, respectively inhibited cell prolifera- and Griffin-Thomas, 2008; Kreitzer and Stella, 2009) and in some
tion and migration in breast cell lines. The results were in line with benign paediatric astrocyte tumours (Ellert-Miklaszewska et al.,
in vivo findings where in mammary gland tumours in the polyoma 2007). The anti-tumour effect of cannabinoids on gliomas, glio-
middle T onco-protein (PyMT) transgenic mice model, a geneti- blastoma multiforme or astrocytoma that are the most frequent
cally engineered model of triple negative breast cancer, WIN- class of malignant primary brain tumours, will be discussed below.
55,212–2 or JWH-133 showed a reduction in tumour growth and Although the downstream events by which cannabinoids exert
lung metastasis. Inhibition induced by the agonists was sensitive their action are not completely elucidated, it can be generally as-
to antagonism by CB1 and CB2 antagonists AM 251 and SR144528, sumed that they act at least through two mechanisms: induction
suggesting involvement of CB1 and CB2 receptors. of apoptosis of tumour cells and/or inhibition of tumour angio-
In terms of signal transduction pathways, cyclooxygenase-2 genesis and migration.
and prostaglandin E2, via the regulation of GTPases and tran- The anti-tumour action of two cannabinoid receptor agonists,
scription factors, have been implicated in the action of cannabi- Δ9-THC and WIN-55,212–2, was shown to be mediated by an in-
noids on breast cancer growth and metastasis (Qamri et al., 2009). crease in the level of ceramide leading to the activation of extra-
Such studies also reported a significant reduction in angiogenesis cellular signal-regulated kinase (ERK1/2) in C6 glioma cells (Galve-
(CD31 staining) and attenuation of proliferation (Ki67 staining). A Roperh et al., 2000; Guzmán, 2003). In addition, positive actions of
reduction in the level of Cdc42 activity and nuclear expression of CP 55–940 include a selective induction of cell death in a hybrid
transcription factors c-Jun and c-Fos in MDA–MB231 cells were cell line of neuroblastoma plus glioma where consistent anti-
also reported (Qamri et al., 2009). proliferative effects were observed (Tomiyama and Funada, 2011).
The involvement of CB1 as well as TRPV1 receptors on the in- It was of interest to note that CP 55–940 had a selective and dif-
vasiveness of MDA-MB-231 cells has been recently discussed. Se- ferential action on C6 compared to the U373 astrocytoma cell lines.
lective agonists were shown to reduce cell invasion and accord- Results showed that C6 cells were dying faster than the U373 cells
ingly MMP-2 expression (Farsandaj et al., 2012). In addition, a mainly via necrotic mechanisms after being exposed to CP 55–940,
down-regulation of vascular endothelial growth factor and con- whilst U373 cells underwent early apoptosis and displayed a more
comitantly over-expression of COX-2 were also reported (Farsandaj defined laddering pattern (Ortega et al., 2015). This illustrates that
et al., 2012). In addition, phytocannabinoids were shown to be as the effects of cannabinoids on cancer cell lines in vitro is context
effective as synthetic compounds (Ligresti et al., 2006). Particu- specific.
larly, CBD was reported to be effective at inhibiting cell prolifera- Other studies (Ellert-Miklaszewska et al., 2005) also showed a
tion of both hormone-sensitive (MCF-7 ) and hormone-negative down regulation of phosphoinositide 3-kinase (PI3K), protein ki-
(MDA-MB-231) cells showing a combination of cell type depen- nase B (Akt), and ERK signalling pathways, and activated pro-
dent mechanisms of action which include either direct or indirect apoptotic function of Bad protein, which lead to induction of
activation of CB2 and TRPV1 receptors and induction of oxidative apoptosis. In similar experiments using the CB2 receptor agonist
F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14 7

JWH-133, apoptosis was induced in glioma cells via enhanced receptors to mediate an anti-tumour activity. This is in line with
ceramide synthesis de novo (Sánchez et al., 2001a; Sarfaraz et al., studies by Sánchez et al in 2001 who reported a considerable re-
2008). However, in addition to ceramide pathway, other pathways gression of malignant glioma cells by the local administration of a
such as stress-regulated protein p8 which leads to the activation of CB2 selective agonist (Sánchez et al., 2001a). In such studies ad-
activating transcription factor-4 (ATF-4) and cell death-inducible ministration of JWH-133 to an immunocompromised mice model,
kinase (TRB3) were shown as mechanism/s of the anti-tumour Rag-2  /  mice inoculated with rat glioma C6-cells caused a 71%
action of cannabinoids (Carracedo et al., 2006a). Other studies reduction in the tumour growth as compared to the control group
have demonstrated that CBD, via receptor-independent manner, and was found to be prevented by co-administration of the CB2
triggers apoptosis of human glioma cells by a cellular mechanism receptor antagonist, SR144528 but not the CB1 receptor antagonist,
that involves an early production of reactive oxygen species (ROS), SR141716. Further experiments by the same group indicated that
depletion of glutathione (GSH), and concomitant activation of the anti-tumour effects induced by the activation of CB2 receptors
caspase cascade with no effect in non-transformed cells (Massi by JWH-133 initiated apoptosis via ceramide synthesis and ERK1/2
et al., 2006). Moreover, the same group, reported that CBD can activation (Sánchez et al., 2001a). In later studies by Blazquez and
inhibit proliferation and invasion of different glioma cell lines colleagues in 2003 using the same mouse model, intra-tumour
through a multi-target mechanism affecting the most relevant administration of JWH-133 showed a significant reduction in
pro-tumour ERK and PI3K/Akt signalling pathways, as well as the mRNA expression of the pro-angiogenic factors, vascular en-
expression of the transcription factor HIF-1α which was down dothelial growth factor and angiopoietin 2 (Blazquez et al., 2003).
following a treatment with CBD. Such experiments were con- Additional data to support the importance of CB2 receptors in
ducted under ‘pseudo-hypoxic conditions’ (Solinas et al., 2013). glioblastoma came from studies that showed an increase in the
This raises the possibility that these compounds could have ac- level of CB2 receptors in the endothelial cells of human glio-
tivity against hypoxic cells but to the best of our knowledge, no blastoma vessels (Schley et al., 2009). Whilst the mechanism of
studies of this nature have been conducted. action of CB agonists against glioblastoma is still not fully under-
Remarkably, the cannabinoid-mediated anti-proliferative ac- stood, it has been shown in the rat C6 glioma cells that the high
tion appears to be selective for brain-tumour cells as the survival affinity glycine transporter (GLYT1) has been attenuated via pro-
of normal brain cells astrocytes (Gomez Del Pulgar et al., 2002), tein kinase C alpha (PKC-α) (Morioka et al., 2008). Attenuation of
oligodendrocytes (Molina-Holgado et al., 2002) and neurons GLYT1 would increase the inhibitory action of glycine transmitter
(Mechoulam et al., 2002) are unaffected or even favoured by in the synapse. Other studies showed that PKC inhibitors could
cannabinoid challenge. Accordingly, a pilot phase I clinical trial, in impair the CB effects in neuroblastoma cells (Rubovitch et al.,
a cohort of recurrent glioblastoma multiforme tumour patients 2004). The above data suggest the involvement of downstream
expressing cannabinoid receptors reported an anti-proliferative PKC and GLYT1 regulation in mediating an anti-glioblastoma ac-
action for Δ9-THC on tumour cells with an acceptable safety profile tivity by cannabinoids. Thus, the accumulated data suggest that
(Guzman et al., 2006). Further phase 1b/2a clinical trials are un- the anti-tumour activity of cannabinoids could be mediated via
derway and await publication. ERK1/2, AKt and/or PI3K pathways as well as PKC and glycine
Based on these findings, other studies evaluated the expression transporters. The change in the levels of cannabinoid receptor
of cannabinoid receptors in surgical material of solid astrocytomas, expression and the levels of endocannabinoids depend on the
gliomas and glioma cell lines. Whilst CB1 expression was slightly stages of cancer. For example, the levels of CB2 receptors and
increased in astrocytomas and gliomas, the CB2 expression was anandamide increase in advanced tumours.
similar in both tumour and normal brain tissue. The authors found,
in accordance with this receptor subtype expression in situ, that
agonists selective for CB1 or active on both subtypes reduced 5. Cannabinoid and lung cancer
elevated cyclic AMP levels and cell proliferation, but failed to in-
duce apoptosis in glioma cells in vitro (Held-Feindt et al., 2006). The first evidence of the antineoplastic activity of cannabinoids
Further experiments described opposite changes in CB1 and CB2 against lung cancer dates back to 1975 when Munson et al de-
receptor protein expression in human gliomas (López-Moreno monstrated a dose-dependent retardation in tumour growth in the
et al., 2010). The reduction in the level of CB1 receptor expression Lewis lung adenocarcinoma animal model (Munson et al., 1975).
or mRNA in glial tumours is suggested to be related to the neu- Later on, further studies were carried out in order to elucidate the
ronal loss (Canoll and Goldman, 2008). The reduction in CB1 re- possible mechanism of action(s) of this class of molecule although
ceptor expression was in line with a reduction in the WIN 55,212– controversial evidence about the anti-tumour action of cannabi-
2 stimulated [35S]GTPγS binding to glioblastoma multiforme noids were reported for this particular type of cancer. In fact, it has
membranes. This suggested a reduction in the number of available been reported that Δ9-THC suppresses the host immune reactivity
receptors in the glioblastomas. Similar observations were reported against lung cancer and that the augmentation of tumour growth
in the brain of aged rats (Romero et al., 1998). It should be noted acts through inhibition of anti-tumour immunity by a CB2 re-
that similar reductions were also observed in neurodegenerative ceptor-mediated, cytokine-dependent pathway (Zhu et al., 2000).
diseases such as Alzheimer, Parkinson and Huntington diseases, Δ9-THC, although without the modulation of EGFR expression or
and also in normal aging (Glass et al., 1993; Hurley et al., 2003; FAK phosphorylation previously reported by others (Hart et al.,
Richfield and Herkenham, 1994; Westlake et al., 1994). In parti- 2004), has been also shown to attenuate the EGF-induced migra-
cular the up-regulation of CB2 receptors has also been reported in tion and invasion of epidermal growth factor receptor-over-
other disorders such as Alzheimer, Huntington diseases, en- expressing lung cancers, which are often highly aggressive and
cephalitis and multiple sclerosis (Benito et al., 2007, 2003; Fer- resistant to chemotherapy (Preet et al., 2008). Furthermore, in
nandez-Ruiz et al., 2007). It is suggested that reduction in the level in vivo experiments, administration of Δ9-THC suppressed metas-
of CB1 receptors occurs as a result of neuronal loss, possibly si- tasis and subcutaneous tumour growth in severe combined im-
multaneous to the enhanced brain gliosis that appears with nor- munodeficient mice (Preet et al., 2008). The anti-invasive effect of
mal aging. In addition to an up-regulation of CB2 receptors in cannabinoids has also been reported (Ramer and Hinz, 2008; Ra-
malignant tumours, the level of anandamide was reported to in- mer et al., 2010). In these studies, a cannabinoid receptor and
crease in glioblastomas (Petersen et al., 2005). It can then be TRPV1-triggered expression of tissue inhibitor of matrix metallo-
suggested that the endogenous anandamide will bind to CB2 proteinases-1 (TIMP-1) was identified as an important mediator of
8 F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14

the anti-invasive action of cannabidiol. Impaired invasion driven role in the regulation of intestinal tumour growth. Recent studies
by CBD links the cannabinoid and TRPV1 receptors to the activa- have demonstrated an up-regulation of anandamide and its me-
tion of MAPK pathways and subsequent TIMP-1 induction. Ad- tabolite arachidonic acid in cancer tissues of patients with colon
ditionally and in line with its in vitro anti-invasive action, in vivo cancer with lymphatic metastasis (Chen et al., 2015). In addition,
studies in thymic-aplastic nude mice revealed a significant in- CB1 receptor expression was elevated (Chen et al., 2015). Another
hibition of A549 lung metastasis in cannabidiol-treated animals as recent study also demonstrated that the level of expression of CB2
compared to vehicle-treated controls (Ramer and Hinz, 2008; receptors correlates with cancer progression and can predict pa-
Ramer et al., 2010). tient survival in colon cancer patients. Such studies showed that
The expression of CB1 and CB2 receptors has been reported in high levels of CB2 receptors correlate with poor prognosis in pa-
non-small cell lung cancer (NSCLC) patients and also the NSCLC tients with tumours in advanced stages or with vascular invasion
cell lines, A549 and SW-1573 (Preet et al., 2011). Pre-treatment of (Martinez-Martinez et al., 2015).
A549 and SW-1573 cells with WIN 55,212–2 and JWH-015 reduced There are however several controversies regarding the role of
chemotaxis and chemo-invasion as well as migration and these the cannabinoid receptors in colorectal cancer (Izzo and Camilleri,
were sensitive to blockade by the CB1 and CB2 receptor antago- 2009). In adenomatous polyposis coli (APC) gene knock-out
nists, AM251 and AM630 respectively. Furthermore both agonists models (mutation of the gene leads to colon cancer), mice with an
revealed a reduction in tumour growth in vitro, inhibited in vivo additional deletion in the cannabinoid receptor 1 (CNR1) gene or
tumour growth and lung metastasis that again were sensitive to subjected to pharmacological blockade of the CB1 receptor, de-
antagonism of CB1 and CB2 receptors (Preet et al., 2011). Me- monstrated a higher colonic tumour burden than their littermates
chanistic studies revealed an inhibition of phosphorylation of Akt whereas activation of CB1 attenuated intestinal tumour growth by
and MMP-9 expression/activity upon exposure to the CB1 and CB2 inducing cell death via down-regulation of the anti-apoptotic
receptor agonists in NSCLC (Preet et al., 2011). On the other hand, factor survivin (Wang et al., 2008). In contrast to these findings,
the involvement of cannabinoid receptor is not the only me- the CB1 antagonist rimonabant inhibited the growth of cancer cells
chanism through which cannabinoids exhibit their anti-tumour and the development of precancerous lesions in mice (Santoro
proprieties. In fact, as demonstrated by Gardner et al. these mo- et al., 2009). Other studies showed that non-selective cannabinoid
lecules can exert some of their biological effects via modulation of receptor agonists such as anandamide, 2-AG and HU-210, and an
prostaglandin production. This study has shown that administra- inhibitor of anandamide inactivation, potently inhibited human
tion of methanandamide at 5.0 mg kg  1 in murine lung cancer epithelial colorectal adenocarcinoma cells (CaCo-2 cell) prolifera-
could increase the rate of tumour growth (both in vitro and tion (Ligresti et al., 2003). This effect was less prominent in a less
in vivo). Such studies also showed an increase in the level of aggressive human colon carcinoma cell line (DLD-1 cells). The cell
prostaglandin (PG) E2 and COX-2 which were sensitive to blockade proliferation effect afforded by HU-210 was inhibited by the CB1
by the COX-2-specific inhibitor, SC58236, the p38/MAPK inhibitor, and CB2 receptor antagonists, rimonabant and SR144528 respec-
SB203528, and the p42/44 inhibitor, PD98059 but not to CB1 and tively, only in DLD-1 cells and not in CaCo-2 cells. This suggested
CB2 receptor antagonists. The results confirmed an up-regulation the involvement of both CB1 and CB2 receptors in mediating an
of COX-2 which was independent of cannabinoid receptor activa- inhibition of cell growth (Ligresti et al., 2003). The differential
tion (Gardner et al., 2003). Recently studies have shown an up- effects observed in the two different cell lines might be due to
regulation of COX-2 induced by CBD in two different lung cancer differences in the level of cannabinoid receptor expression. Indeed,
cell lines (A549, H460) and primary cells from a patient with lung it was noted that CaCo-2 cells express CB1 receptors but not CB2
cancer (Ramer et al., 2013). Such studies showed a pro-apoptotic receptors and DLD-1 cells express both CB1 and CB2 receptors,
and tumour-regressive action by CBD through an initial up-reg- with CB1 receptor less expressed than in CaCo-2 cells. Later studies
ulation of COX-2 and PPAR-γ and a subsequent nuclear transloca- suggested that cells with high expression of cyclooxygenase-2
tion of PPAR-γ by COX-2-dependent prostaglandins, PGD2 (Ramer (COX-2) might be a target for the inhibitory action of anandamide
et al., 2013). Such studies also indicated that incubation of the cells on cell death in colorectal carcinoma cells (Patsos et al., 2005).
with PGD2 but not PGE2 was associated with a concentration- Indeed CB2 receptor expression has been reported in human
dependent loss of cell viability. This highlighted how different adenomatous polyps and carcinomas and in human colonic epi-
cannabinoids could induce differential effects, which are CB1 and thelial cell lines (Greenhough et al., 2007; Ihenetu et al., 2003;
CB2 receptor-independent pathways and also dependent on the Ligresti et al., 2003; Wright et al., 2005). Interestingly the normal
type of PG being produced. epithelial cells do not express CB2 receptors (Wright et al., 2005).
In further recent studies, it was demonstrated that cannabi- This suggests that the CB2 receptors are inducible in inflamed
noids induced up-regulation of intercellular adhesion molecule 1 tissues or tumour cells. This suggestion is in line with other studies
(ICAM-1) on lung cancer cells to be responsible for increased which reported an increase in the level of CB2 receptor expression
cancer cell lysis by lymphokine-activated killer (LAK) cells (Haus- associated with increased differentiation, proliferation, disease
tein et al., 2014). This recent study suggested a new mechanism for and malignancy (Fernandez-Ruiz et al., 2007; Mallat and Lo-
anti-tumour activity of cannabinoids. Further experiments by Ra- tersztajn, 2008). Ceramide synthesis following an increase in the
mer et al also indicated that cannabinoids induce anti-angiogenic level of tumour necrosis factor (TNF)-α and activation of epidermal
effect in endothelial cells via the release of TIMP-1 from lung growth factor receptor have been identified as the possible mo-
cancer cells (Ramer et al., 2014). lecular mechanism following the activation of CB2 receptors in
Thus, the likely pathways involved in mediating anti-tumour colon cancer (Cianchi et al., 2008; Hart et al., 2004). Thus since
activity induced by cannabinoids are CB1, CB2 and TRPV1 receptors activation of CB2 receptors seem to be beneficial in cancer therapy,
as well as MAPK and TIMP-1 pathways. However, this does not further studies are needed to investigate the dual role for CB2
negate a role for PPAR-γ and COX-2-dependent prostaglandins. receptors in intestinal regeneration and anti-tumour activity. It is
also possible that different subtypes of CB2 receptors mediate
different roles, however no evidence has yet been reported.
6. Cannabinoids and intestinal cancer Cannabinoids have been also reported to exert chemopreven-
tive effects in an experimental model of colon cancer, an effect
Endocannabinoid signalling has been proved crucial for certain associated with down-regulation of phospho-Akt and up-regula-
aspects of gastrointestinal homoeostasis. It also plays an essential tion of caspase-3 (Aviello et al., 2012). In vitro studies by the same
F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14 9

group on colorectal carcinoma cells demonstrated that anti-pro- complete endogenous machinery for CB2 activation was altered in
liferative effects were exerted through multiple mechanisms, in- endometrial adenocarcinoma, because the levels of 2-AG, the most
cluding involvement of CB1 receptors, TRPV1 and PPAR-γ (Aviello efficacious endogenous CB2 agonist, were elevated, possibly as a
et al., 2012). Moreover, studies by Notarnicola and collaborators result of the decrease in the expression of monoglycerol lipase
firstly described the up-regulation of CB1 expression by 17β-es- (MAGL) an important enzyme necessary for 2-AG breakdown. On
tradiol as a further mechanism by which estrogens control colon the other hand, CB1 receptors and AEA, a more selective en-
cancer cell proliferation (Notarnicola et al., 2008). Proto and col- dogenous agonist for CB1, as well as FAAH, the most important
leagues confirmed these findings and reported an interaction be- AEA-metabolising enzyme, although expressed in healthy en-
tween the endocannabinoid system and steroid hormones in the dometrial tissues, remained unchanged after cell transformation
growth of colon cancer cells. Such studies revealed that both (Guida et al., 2010).
anandamide and 17β-estradiol inhibited proliferation of human Additional proteins other than cannabinoid receptors have
colorectal cancer cell lines, SW620 and DLD-1 via interaction with been considered as possible targets in reproductive cancers. Mul-
CB1 receptors. Both agonists increased the CB1 receptor expression tidrug resistance-related protein 1 (MRP1) or ATP-binding cassette
in both cell lines by acting at the same CNR1 gene. Interestingly (ABC) transporter, ABCC1, is a membrane-bound, ubiquitously
the up-regulation of CB1 receptors induced by anandamide ana- expressed energy-dependent efflux transporter. In terms of phy-
logueue was through PPAR-γ and RXRα, (Proto et al., 2012). The siological function, it is involved in transporting a range of glu-
data suggested that CB1 receptor is a target for 17β-estradiol and tathione, glucuronide, sulphate conjugates and cancer drugs, in-
that the endocannabinoid system could present a tool to improve cluding folate based anti-metabolites, anthracyclines, plant-de-
treatment in patients with colorectal cancer. rived vinca alkaloids and anti-androgens (Cole et al., 1994; Flens
et al., 1996; Hooijberg et al., 1999; Keppler et al., 1997). Whilst the
transportation of metabolites by ABCC1 leads to attenuation of the
7. Cannabinoids and reproductive system cancer toxicity of such metabolites might be beneficial, efflux of cancer
drugs would however reduce intracellular concentrations in tu-
During the last decade, increasing evidence has pointed to- mour cells and hence induce drug resistance (Karászi et al., 2001;
wards the relevance of endocannabinoids in both female and male Norris et al., 1996; Wijnholds et al., 2000, 1998). It has been shown
fertility. This association has been supported by the tightly that phytocannabinoids are modulators of the ABC transporters,
modulated expression of cannabinoid receptor found in gonadal ABCG2 and P-glycoprotein (Holland et al., 2007; Zhu et al., 2000).
tissues. Along the male reproductive tract, CB receptors have been It was also shown in the human ovarian carcinoma cell line that
detected in the testis, Sertoli cells, prostate and vas deferens (Gye cannabinoids such as cannabinol, cannabidiol and Δ9-THC in-
et al., 2005; Maccarrone et al., 2003; Pertwee et al., 2002; Rossato creased the intracellular accumulation of two ABCC1 substrates,
et al., 2005). CB receptors have also been found in various parts of Fluo3 and the cancer drug, vincristine, in 2008/MRP1 cells (the
the mammalian female reproductive system. In the mouse re- human ABCC1 transduced subline) (Holland et al., 2008). In such
productive tract, CB receptors were expressed in the uterus, ovi- experiments cannabidiol was shown to be the most potent and Δ9-
duct and also in pre-implantation embryos (Das et al., 1995; Paria THC to be the least potent cannabinoid. The rank order of potency
et al., 2001; Wang et al., 2004). Moreover, this localisation has also for ABCC1 inhibition was independent of the substrate assayed
been described in the human uterus (Dennedy et al., 2004; Iuvone (Holland et al., 2008). Further pre-clinical studies are required to
et al., 2008) and placenta during pregnancy (Habayeb et al., 2008; establish if inhibition of ABC transporters by cannabinoids can
Helliwell et al., 2004; Park et al., 2003). alter the disposition and efficacy of therapeutic drugs that are
The influence of cannabinoids on the proliferation of human substrates for these transporters.
cervical adenocarcinoma cells and on macromolecular biosyn- With regard to the male reproductive system and its physiol-
thetic events associated with the proliferative process were re- ogy, the antagonising effect of cannabinoids can be dated back to
ported in the late 1970s when cannabinoids induced growth in- 1974 where experimental models in male rats showed depression
hibition of HeLa cells, human cervical cancer cell lines (Blevins of spermatogenesis (Dixit et al., 1974) and decrease in circulating
et al., 1980; Mon et al., 1978). More recently, an up-regulation of testosterone levels (Kolodny et al., 1974). Endocannabinoids,
both cannabinoid receptors in human ovarian cancer cells OVCAR- through interaction with CB1 receptors and synthetic en-
3 and SKOV-3 compared to normal Chinese hamster ovarian (CHO) docannabinoid-vanilloid hybrids via stimulation of TRPV1 chan-
cells was found. These findings led to the suggestion that these are nels have been shown to inhibit nerve growth factor (NGF)-in-
targets for new therapies for ovarian cancer (Afaq et al., 2006). duced proliferation of human prostate PC-3 cells (Melck et al.,
Recently studies also showed that CB1 receptor levels are also in- 2000). However, THC was suggested to induce apoptosis of these
creased and correlate with disease severity in human epithelial cells via a receptor-independent mechanism (Ruiz et al., 1999), but
ovarian tumours and this has been proposed to be an important also increase the production of the pro-proliferative factor, NGF
factor of bad prognosis following surgery in stage IV colorectal (Velasco et al., 2001). Later studies showed an increased expres-
cancer (Messalli et al., 2014; Jung et al., 2013). WIN-55,212–2 was sion of both CB1 and CB2 receptors in cultured prostate cancer cells
shown to exert, via CBR dependent manner, a decrease in cell when compared with normal prostate cells. Moreover, treatment
viability, G1 arrest in cell cycle progression, induction of apoptosis of prostate cancer cells with WIN-55,212–2 resulted in a dose and
and down-regulation of the expression of PCNA and VEGF (Afaq time dependent decrease in cell viability, increased apoptosis
et al., 2006). An abnormal expression of CB2 receptor has also been along with decrease in androgen receptor protein expression, PSA
reported in biopsies of women affected by endometrial carcinoma. expression, and secreted PSA, suggesting that cannabinoids should
Interestingly, the up-regulation was only found in transformed be considered as agents for the management of prostate cancer
malignant cells and the staining of CB2 was completely absent in (Ruiz-Llorente et al., 2003; Sanchez et al., 2003; Sarfaraz et al.,
the normal endometrial tissue from the same biopsy (Guida et al., 2005). It was also found that a high level of CB1 receptor im-
2010). These findings, together with previous evidence that the munoreactivity (CB1IR) in prostate cancer tissues is associated
endocannabinoid system controls cell survival/death decisions with the severity and outcome of the disease (Chung et al., 2009).
(Guzman et al., 2002) by inhibiting or stimulating cell growth, It was then suggested that the effect of cannabinoids on pros-
suggest that CB2 receptors might play an important role in the tate cancer cells depends on the concentration of cannabinoids
growth of endometrial carcinoma. The study revealed that the used and the incubation time. Cannabinoids used at
10 F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14

concentrations lower than micromolar induced androgen receptor 8. Conclusion and future direction
expression whilst at higher concentrations induced apoptosis or
cell-cycle arrest (Mimeault et al., 2003; Sanchez et al., 2003; Sar- The substantial knowledge of palliative and anti-tumour ac-
faraz et al., 2006). It was also shown that CB1 receptor antagonists tions of cannabinoids gained by the scientific community in the
prevented/blocked the anti-tumour activity of agonists whilst a last few years has raised the profile of these molecules and many
longer incubation time failed to reveal any antagonist effect (Mi- are promising candidates for cancer treatment. However, the use
meault et al., 2003; Sarfaraz et al., 2005). of cannabinoids in medicine is limited by their psychoactive ef-
Other studies have also indicated an important role for CB2 but fects, thus cannabinoid-based therapies that are devoid of un-
not CB1 receptors in the anti-proliferative effect of cannabinoids wanted side effects or with a safe profile/pharmacological window
(Olea-Herrero et al., 2009). Such studies have shown that in PC-3 are required. A further aspect which complicates the practise of
cells pre-treated with rimonabant failed to reduce the effect of cannabinoid-based therapies is the lack of detailed understanding
methanandamide on cell cycle and apoptosis. However, pre- of their mechanisms of action. There is plenty of evidence in lit-
treatment with the CB2 receptor antagonist, SR 144528 attenuated erature, mostly of them reported in this review, about the ability of
the number of apoptotic cells and the number of sub-G1 cells in- cannabinoids to induce different pathways of cell death depending
duced by methanandamide and apoptosis afforded by the CB2 on the neoplastic cell type under investigation. On the other hand,
receptor agonist, JWH-015. Furthermore, when CB2 receptor ex- the effect of these compounds on several distinct hallmarks of
pression was significantly reduced by siRNA, apoptosis afforded by cancer rather than on one single process is potentially desirable.
JWH-015 was completely antagonised thus further indicating an Even though the resolution of the conflicting evidence around
involvement of CB2 receptors in apoptosis (Olea-Herrero et al., cannabinoid action still remains a high research priority, some key
2009). points need to be emphasised. Despite a small number of reports
Recent studies have focused on the role of non-selective, cal- that state their inefficacy, the vast majority of independent pre-
cium permeable cation channels of the transient receptor potential clinical studies report a sustained anti-tumour activity for canna-
(TRP) channels in prostate cancer initiation and progression. binoids. Moreover, an important feature of cannabinoid pharma-
Capsaicin, a natural ligand for TRPV1, has been reported to elicit cology that can have important clinical implications is the lack of
both pro-proliferative and pro-apoptotic effects on prostate cancer toxicity frequently reported on non-tumour cells. Cannabinoid-
cell lines (Czifra et al., 2009; Malagarie-Cazenave et al., 2009, 2011; based medicines have been already proven to be safe in thousands
Sánchez et al., 2006, 2005; Ziglioli et al., 2009). Moreover, it has of patients enroled in clinical trials for cancer patients (Gro-
been suggested that TRP channels of melastatin-type 8 (TRPM8) tenhermen, 2007; Portenoy et al., 2012; Robson, 2011). This
are over-expressed in androgen-dependent prostate cancer cell highlights a need for the identification of the molecular mechan-
lines in a manner dependent on androgen receptor (AR) activation isms which confer sensitivity to this class of drugs. Another ap-
(Bidaux et al., 2007, 2005; Henshall et al., 2003; Tsavaler et al., pealing possibility consists of adding to a standard therapy a
2001; Zhang and Barritt, 2004). Several studies have shown that mixture of molecules able to directly or indirectly target the en-
cannabinoids antagonise TRPM8 channels and activate and sub- docannabinoids system in order to enhance anti-tumour actions of
sequently desensitise TRPV2 and TRPV1 channels (De Petrocellis chemotherapy and attenuate unwanted iatrogenic side effects.
et al., 2011, 2008; Qin et al., 2008). Further recent studies showed Indeed it is interesting to note that the administration of THC and
an inhibition of cell viability and induction of apoptosis by can- CBD enhanced the radio-therapeutic effect in an orthotopic mur-
nabinoids when tested in serum protein-deprived medium sug- ine glioma model (Scott et al., 2014). It is proposed that CBD could
gesting an intracellular target for the cannabionids (De Petrocellis alleviate the THC-induced side effects such as convulsion, dis-co-
et al., 2013). However, the molecular mechanism of action was ordination, and psychotic episodes, thus it was suggested that the
demonstrated to be due not uniquely to a direct TRPM8 antag- administration of CBD in combination with THC may help to im-
onism, but rather to AR down regulation, which in turn can lead to prove the tolerability to cannabinoids (Pertwee, 2009). Finally,
TRPM8 down-regulation. As it has been suggested that estrogens only the improvement of basic research will lead to the identifi-
are involved in the survival of prostate cells, the authors also ex- cation of the most appropriate patient population for a cannabi-
amined the involvement of those receptors and they found that noid-based therapy and will facilitate the acceptance of cannabi-
GPR30, rather than estrogen metabolic enzymes or ERα or ERβ, noid use in the clinic.
may be one of the intracellular targets through which cannabi-
noids stimulate the ER branch of the intrinsic pro-apoptotic
pathway Δ9-THC (De Petrocellis et al., 2013). Conflict of interests/acknowledgement
In addition, the expression of a potential cannabinoid receptor,
GPR55 has been also shown at the mRNA and protein level in both FJ and AL disclose that they have received research support
ovarian and prostate cancer cell lines (Pineiro et al., 2011). Al- funding from GW Pharma.
though the physiological role of this receptor is not fully under-
stood, it is suggested to have important roles in regulating pro-
liferation and growth (Pineiro et al., 2011). This novel receptor is References
suggested to be associated with lysophosphatidylinositol (LPI) to
induce calcium mobilisation and activation of Akt and extracellular Afaq, F., Sarfaraz, S., Syed, D., Khan, N., Malik., A., Bailey, H., et al., 2006. Cannabinoid
signal-regulated kinase (ERK)1/2 in ovarian and prostate cell lines. Receptors as a Target for Therapy of Ovarian Cancer. AACR Meeting, Wa-
shington, DC.
Blockade of this receptor might have novel therapeutic potential in Ameri, A., 1999. The effects of cannabinoids on the brain. Prog. Neurobiol. 58 (4),
managing ovarian and prostate cancer. All these findings indicate 315–348.
that cannabinoids can retard proliferation and cause apoptosis of Andradas, C., Caffarel, M.M., Perez-Gomez, E., Salazar, M., Lorente, M., Velasco, G.,
et al., 2011. The orphan G protein-coupled receptor GPR55 promotes Cancer cell
PCC via a combination of cellular and molecular mechanisms
proliferation via ERK. Oncogene 30 (2), 245–252.
(cannabinoid receptor-mediated and/or independent). These stu- Armstrong, J.L., Hill, D.S., McKee, C.S., Hernandez-Tiedra, S., Lorente, M., Lopez-
dies are encouraging and they support the development of clinical Valero, L., et al., 2015. Exploiting cannabinoid-induced cytotoxic autophagy to
studies on these molecules as a therapy for human prostate car- drive melanoma cell death. J. Invest. Dermatol. 135 (6), 1629–1637.
Aviello, G., Romano, B., Borrelli, F., Capasso, R., Gallo, L., Piscitelli, F., et al., 2012.
cinoma, either as single agent or in combination with existing Chemopreventive effect of the non-psychotropic phytocannabinoid cannabidiol
compounds. on experimental colon Cancer. J. Mol. Med. 90 (8), 925–934.
F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14 11

Benito, C., Nunez, E., Pazos, M.R., Tolon, R.M., Romero, J., 2007. The en- cell proliferation. Proc. Natl. Acad. Sci. USA 95 (14), 8375–8380.
docannabinoid system and Alzheimer disease. Mol. Neurobiol. 36 (1), 75–81. De Petrocellis, L., Vellani, V., Schiano-Moriello, A., Marini, P., Magherini, P., Orlando,
Benito, C., Nunez, E., Tolon, R.M., Carrier, E.J., Rabano, A., Hillard, C.J., et al., 2003. P., et al., 2008. Plant-derived cannabinoids modulate the activity of transient
Cannabinoid CB2 receptors and fatty acid amide hydrolase are selectively receptor potential channels of ankyrin type-1 and melastatin type-8. J. Phar-
overexpressed in neuritic plaque-associated glia in Alzheimer disease brains. J. macol. Exp. Ther. 325 (3), 1007–1015.
Neurosci. 23 (35), 11136–11141. De Petrocellis, L., Ligresti, A., Schiano Moriello, A., Allarà, M., Bisogno, T., Petrosino,
Benz, A.H., Renne, C., Maronde, E., Koch, M., Grabiec, U., Kallendrusch, S., et al., 2013. S., et al., 2011. Effects of cannabinoids and cannabinoid-enriched cannabis ex-
Expression and functional relevance of cannabinoid receptor 1 in Hodgkin tracts on TRP channels and endocannabinoid metabolic enzymes. Br. J. Phar-
lymphoma. PLoS One 8, e81675. macol. 163 (7), 1479–1494.
Bidaux, G., Roudbaraki, M., Merle, C., Crépin, A., Delcourt, P., Slomianny, C., et al., De Petrocellis, L., Ligresti, A., Schiano Moriello, A., Iappelli, M., Verde, R., Stott, C.,
2005. Evidence for specific TRPM8 expression in Human prostate secretory et al., 2013. Non-THC cannabinoids inhibit prostate carcinoma growth in vitro
epithelial cells: functional androgen receptor requirement. Endocr. Relat. Can- and in vivo: pro-apoptotic effects and underlying mechanisms. Br. J. Pharmacol.
cer 12 (2), 367–382. 168 (1), 79–102.
Bidaux, G., Flourakis, M., Thebault, S., Zholos, A., Beck, B., Gkika, D., et al., 2007. Demuth, D.G., Molleman, A., 2006. Cannabinoid signalling. Life Sci. 78 (6), 549–563.
Prostate cell differentiation status determines transient receptor potential Dennedy, M., Friel, A., Houlihan, D., Broderick, V., Smith, T., Morrison, J., 2004.
melastatin member 8 channel subcellular localization and function. J. Clin. In- Cannabinoids and the Human uterus during pregnancy. Am. J. Obstet. Gynecol.
vest. 117 (6), 1647–1657. 190 (1), 2–9.
Bifulco, M., Laezza, C., Portella, G., Vitale, M., Orlando, P., De Petrocellis, L., Di Marzo, Devane, W.A., Hanus, L., Breuer, A., Pertwee, R.G., Stevenson, L.A., Griffin, G., et al.,
V., 2001. Control by the endogenous cannabinoid system of ras oncogene-de- 1992. Isolation and structure of a brain constituent that binds to the cannabi-
pendent tumor growth. FASEB J. 15 (14) 2745-7. noid receptor. Science 258 (5090), 1946–1949.
Bifulco, M., Malfitano, A.M., Pisanti, S., Laezza, C., 2008. Endocannabinoids in en- Di Marzo, V., Bisogno, T., De Petrocellis, L., 2001. Anandamide: some like it hot.
docrine and related tumours. Endocr. Relat. Cancer 15 (2), 391–408. Trends Pharmacol. Sci. 22 (7), 346–349.
Blazquez, C., Casanova, M.L., Planas, A., Gomez Del Pulgar, T., Villanueva, C., Fer- Di Marzo, V., Breivogel, C.S., Tao, Q., Bridgen, D.T., Razdan, R.K., Zimmer, A.M., et al.,
nandez-Acenero, M.J., et al., 2003. Inhibition of tumor angiogenesis by canna- 2000. Levels, metabolism, and pharmacological activity of anandamide in CB
binoids. FASEB J. 17 (3), 529–531. (1) cannabinoid receptor knockout mice: evidence for non-CB(1), non-CB
Blazquez, C., Carracedo, A., Barrado, L., Real, P.J., Fernandez-Luna, J.L., Velasco, G., (2) receptor-mediated actions of anandamide in mouse brain. J. Neurochem. 75
et al., 2006. Cannabinoid receptors as novel targets for the treatment of mel- (6), 2434–2444.
anoma. FASEB J. 20 (14), 2633–2635. Dixit, V.P., Sharma, V.N., Lohiya, N.K., 1974. The effect of chronically administered
Blevins, R., Smith, D., 1980. Effects of delta-9-tetrahydrocannabinol on cultured cannabis extract on the testicular function of mice. Eur. J. Pharmacol. 26 (1),
HeLa cell growth and development. Growth 44, 133–134. 111–114.
Blevins, R.D., Smith, D.P., 1980. Effects of delta-9-tetrahydrocannabinol on cultured Ek, S., Hogerkorp, C.M., Dictor, M., Ehinger, M., Borrebaeck, C.A., 2002. Mantle cell
HeLa cell growth and development. Growth 44 (2) 133-8. lymphomas express a distinct genetic signature affecting lymphocyte traffick-
Breivogel, C.S., Griffin, G., Di Marzo, V., Martin, B.R., 2001. Evidence for a New G ing and growth regulation as compared with subpopulations of normal Human
protein-coupled cannabinoid receptor in mouse brain. Mol. Pharmacol. 60 (1), B cells. Cancer Res. 62 (15), 4398–4405.
155–163. Elbaz, M., Nasser, M.W., Ravi, J., Wani, N.A., Ahirwar, D.K., Zhao, H., Oghumu, S.,
Buckley, N.E., 2008. The peripheral cannabinoid receptor knockout mice: an update. Satoskar, A.R., Shilo, K., Carson, W.E., Ganju, R.K., 2015. Modulation of the tumor
Br. J. Pharmacol. 153 (2), 309–318. microenvironment and inhibition of EGF/EGFR pathway: novel anti-tumor
Cabral, G.A., Griffin-Thomas, L., 2008. Cannabinoids as therapeutic agents for ab- mechanisms of Cannabidiol in breast Cancer. Mol. Oncol. 9 (4), 906–919.
lating neuroinflammatory disease. Endocr. Metab. Immune Disord. Drug Tar- Ellert-Miklaszewska, A., Kaminska, B., Konarska, L., 2005. Cannabinoids down-
gets 8 (3), 159–172. regulate PI3K/Akt and erk signalling pathways and activate proapoptotic
Caffarel, M.M., Moreno-Bueno, G., Cerutti, C., Palacios, J., Guzman, M., Mechta- function of bad protein. Cell Signal 17 (1), 25–37.
Grigoriou, F., et al., 2008. JunD is involved in the antiproliferative effect of Ellert-Miklaszewska, A., Grajkowska, W., Gabrusiewicz, K., Kaminska, B., Konarska,
Delta9-tetrahydrocannabinol on Human breast Cancer cells. Oncogene 27 (37), L., 2007. Distinctive pattern of cannabinoid receptor type II (CB2) expression in
5033–5044. adult and pediatric brain tumors. Brain Res. 1137 (1), 161–169.
Caffarel, M.M., Andradas, C., Mira, E., Perez-Gomez, E., Cerutti, C., Moreno-Bueno, G., Farsandaj, N., Ghahremani, M.H., Ostad, S.N., 2012. Role of cannabinoid and va-
et al., 2010. Cannabinoids reduce ErbB2-driven breast Cancer progression nilloid receptors in invasion of Human breast carcinoma cells. J. Environ. Pathol.
through Akt inhibition. Mol. Cancer 9, 196. Toxicol. Oncol. 31 (4), 377–387.
Canoll, P., Goldman, J.E., 2008. The interface between glial progenitors and gliomas. Fernandez-Ruiz, J., Romero, J., Velasco, G., Tolon, R.M., Ramos, J.A., Guzman, M.,
Acta Neuropathol. 116 (5), 465–477. 2007. Cannabinoid CB2 receptor: a new target for controlling neural cell sur-
Carracedo, A., Gironella, M., Lorente, M., Garcia, S., Guzman, M., Velasco, G., et al., vival? Trends Pharmacol. Sci. 28 (1), 39–45.
2006a. Cannabinoids induce apoptosis of pancreatic tumor cells via en- Flens, M., Zaman, G., Van der Valk, P., Izquierdo, M., Schroeijers, A., Scheffer, G.,
doplasmic reticulum stress-related genes. Cancer Res. 66 (13), 6748–6755. et al., 1996. Tissue distribution of the multidrug resistance protein. Am. J. Pa-
Carracedo, A., Lorente, M., Egia, A., Blazquez, C., Garcia, S., Giroux, V., et al., 2006b. thol. 148 (4), 1237–1247.
The stress-regulated protein p8 mediates cannabinoid-induced apoptosis of Galve-Roperh, I., Sanchez, C., Cortes, M., Del Pulgar, T., Izquierdo, M., Guzman, M.,
tumor cells. Cancer Cell 9 (4), 301–312. 2000. Anti-tumoural action of cannabinoids: involvement of sustained cer-
Casanova, M.L., Blazquez, C., Martinez-Palacio, J., Villanueva, C., Fernandez-Acenero, amide accumulation and extracellular signal-regulated kinase activation. Nat.
M.J., Huffman, J.W., et al., 2003. Inhibition of skin tumor growth and angio- Med. 6, 313–319.
genesis in vivo by activation of cannabinoid receptors. J. Clin. Invest. 111 (1), Gaoni, Y., Mechoulam, R., 1964a. Hashish II. the structure and synthesis of canna-
43–50. bigerol, a New hashish constituent. Proc. Chem. Soc., 82.
Chakrabarti, A., Onaivi, E.S., Chaudhuri, G., 1995. Cloning and sequencing of a cDNA Gaoni, Y., Mechoulam, R., 1964b. Isolation, structure, and partial synthesis of an
encoding the mouse brain-type cannabinoid receptor protein. DNA Seq. 5 (6), active constituent of hashish. J. Am. Chem. Soc. 86, 1646–1647.
385–388. Gaoni, Y., Mechoulam, R., 1964c. Hashish II. the structure and synthesis of canna-
Chen, L., Li, Y., Li, L., Qiu, Y., 2015. Endocannabinoid and ceramide levels are altered bigerol, a New hashish constituent. Proc. Chem. Soc., 82.
in patients with colorectal Cancer. Oncol. Rep. 34 (1), 447–454. Gaoni, Y., Mechoulam, R., 1966. Cannabichromene, a new active principle in
Chung, S.C., Hammarsten, P., Josefsson., A., Stattin, P., Granfors, T., Egevad, L., et al., hashish. Chem. Commun. 1, 20–21.
2009. A high cannabinoid CB(1) receptor immunoreactivity is associated with Gardner, B., Zhu, L.X., Sharma, S., Tashkin, D.P., Dubinett, S.M., 2003. Metha-
disease severity and outcome in prostate Cancer. Eur. J. Cancer 45 (1), 174–182. nandamide increases COX-2 expression and tumor growth in murine lung
Cianchi, F., Papucci, L., Schiavone, N., Lulli, M., Magnelli, L., Vinci, M.C., et al., 2008. Cancer. FASEB J. 17 (14), 2157–2159.
Cannabinoid receptor activation induces apoptosis through tumor necrosis Glass, M., Northup, J.K., 1999. Agonist selective regulation of g proteins by canna-
factor alpha-mediated ceramide DE novo synthesis in colon Cancer cells. Clin. binoid CB1 and CB2 receptors. Mol. Pharmacol. 56 (6), 1362–1369.
Cancer Res. 14 (23), 7691–7700. Glass, M., Faull, R.L., Dragunow, M., 1993. Loss of cannabinoid receptors in the
Cole, S., Sparks, K., Fraser, K., Loe, D., Grant, C., Wilson, G., et al., 1994. Pharmaco- substantia nigra in Huntington disease. Neuroscience 56 (3), 523–527.
logical characterization of multidrug resistant MRP-transfected Human tumor Gomez Del Pulgar, T., De Ceballos, M.L., Guzman, M., Velasco, G., 2002. Cannabi-
cells. Cancer Res. 54, 5902–5910. noids protect astrocytes from ceramide-induced apoptosis through the phos-
Czifra, G., Varga, A., Nyeste, K., Marincsák, R., Tóth, B., Kovács, I., et al., 2009. In- phatidylinositol 3-kinase/protein kinase B pathway. J. Biol. Chem. 277 (39),
creased expressions of cannabinoid receptor-1 and transient receptor potential 36527–36533.
vanilloid-1 in Human prostate carcinoma. J. Cancer Res. Clin. Oncol. 135 (4), Greenhough, A., Patsos, H.A., Williams, A.C., Paraskeva, C., 2007. The cannabinoid
507–514. delta(9)-tetrahydrocannabinol inhibits RAS-MAPK and PI3K-AKT survival sig-
Das, S., Paria, B., Chakraborty, I., Dey, S., 1995. Cannabinoid ligand-receptor sig- nalling and induces BAD-mediated apoptosis in colorectal Cancer cells. Int. J.
naling in the mouse uterus. Proc. Natl. Acad. Sci. USA 92 (10), 4332–4336. Cancer 121 (10), 2172–2180.
De Petrocellis, L., Di Marzo, V., 2010. Non–CB1, non–CB2 receptors for en- Grimaldi, C., Pisanti, S., Laezza, C., Malfitano, A.M., Santoro, A., Vitale, M., et al.,
docannabinoids, plant cannabinoids, and synthetic cannabimimetics: focus on 2006. Anandamide inhibits adhesion and migration of breast Cancer cells. Exp.
G-protein-coupled receptors and transient receptor potential channels. J. Cell Res. 312 (4), 363–373.
Neuroimmune Pharmacol. 5 (1), 103–121. Grotenhermen, F., 2007. The toxicology of cannabis and cannabis prohibition.
De Petrocellis, L., Melck, D., Palmisano, A., Bisogno, T., Laezza, C., Bifulco, M., et al., Chem. Biodivers. 4 (8), 1744–1769.
1998. The endogenous cannabinoid anandamide inhibits Human breast Cancer Guida, M., Ligresti, A., De Filippis, D., D'Amico, A., Petrosino, S., Cipriano, M., et al.,
12 F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14

2010. The levels of the endocannabinoid receptor CB2 and its ligand 2-arachi- of vascular endothelial growth factor-mediated angiogenesis in tamoxifen-re-
donoylglycerol are elevated in endometrial carcinoma. Endocrinology 151 (3), sistant breast Cancer cells: role of Pin1 overexpression. Mol. Cancer Ther. 8 (8),
921–928. 2163–2171.
Gustafsson, K., Christensson, B., Sander, B., Flygare, J., 2006. Cannabinoid receptor- Kogan, N.M., 2005. Cannabinoids and Cancer. Mini Rev. Med. Chem. 5 (10),
mediated apoptosis induced by R(+)-methanandamide and Win55,212-2 is 941–952.
associated with ceramide accumulation and p38 activation in mantle cell Kolodny, R.C., Masters, W.H., Kolodner, R.M., Toro, G., 1974. Depression of plasma
lymphoma. MolPharmacol. 70 (5) 1612-20. testosterone levels after chronic intensive marihuana use. N. Engl. J. Med. 290
Guzmán, M., 2003. Cannabinoids: potential anticancer agents. Nat. Rev. Cancer 3 (16), 872–874.
(10), 745–755 (Review). Kreitzer, F.R., Stella, N., 2009. The therapeutic potential of novel cannabinoid re-
Guzman, M., Galve-Roperh, I., Sanchez, C., 2001. Ceramide: a new second mes- ceptors. Pharmacol. Ther. 122 (2), 83–96.
senger of cannabinoid action. Trends Pharmacol. Sci. 22 (1), 19–22. Lehen'kyi, V., Prevarskaya, N., 2011. Oncogenic TRP channels. Adv. Exp. Med. Biol.
Guzman, M., Sanchez, C., Galve-Roperh, I., 2002. Cannabinoids and cell fate. Phar- 704, 929–945.
macol. Ther. 95 (2), 175–184. Liberati, S., Morelli, M.B., Nabissi, M., Santoni, M., Santoni, G., 2013. Oncogenic and
Guzman, M., Duarte, M.J., Blazquez, C., Ravina, J., Rosa, M.C., Galve-Roperh, I., et al., anti-oncogenic effects of transient receptor potential channels. Curr. Top. Med.
2006. A pilot clinical study of Delta9-tetrahydrocannabinol in patients with Chem. 13 (3), 344–366.
recurrent glioblastoma multiforme. Br. J. Cancer 95 (2), 197–203. Ligresti, A., Bisogno, T., Matias, I., De Petrocellis, L., Cascio, M.G., Cosenza, V., et al.,
Gye, M.C., Kang, H.H., Kang, H.J., 2005. Expression of cannabinoid receptor 1 in 2003. Possible endocannabinoid control of colorectal Cancer growth. Gastro-
mouse testes. Arch. Androl. 51 (3), 247–255. enterology 125 (3), 677–687.
Habayeb, O., Taylor, A., Bell, S., Taylor, D., Konje, J., 2008. Expression of the en- Ligresti, A., Moriello, A.S., Starowicz, K., Matias, I., Pisanti, S., De Petrocellis, L., et al.,
docannabinoid system in Human first trimester placenta and its role in tro- 2006. Antitumor activity of plant cannabinoids with emphasis on the effect of
phoblast proliferation. Endocrinology 149 (10), 5052–5060. cannabidiol on Human breast carcinoma. J. Pharmacol. Exp. Ther. 318 (3),
Hart, S., Fischer, O.M., Ullrich, A., 2004. Cannabinoids induce Cancer cell pro- 1375–1387.
liferation via tumor necrosis factor alpha-converting enzyme (TACE/ADAM17)- López-Moreno, J.A., López-Jiménez, A., Gorriti, M.A., de Fonseca, F.R., 2010. Func-
mediated transactivation of the epidermal growth factor receptor. Cancer Res. tional interactions between endogenous cannabinoid and opioid systems: focus
64 (6), 1943–1950. on alcohol, genetics and drug-addicted behaviors. Curr Drug Targets 11 (4) 406-
Haustein, M., Ramer, R., Linnebacher, M., Manda, K., Hinz, B., 2014. Cannabinoids 28. Review.
increase lung Cancer cell lysis by lymphokine-activated killer cells via upre- Maccarrone, M., Lorenzon, T., Bari, M., Melino, G., Finazzi-Agro, A., 2000. Ananda-
gulation of ICAM-1. Biochem. Pharmacol. 92 (2), 312–325. mide induces apoptosis in human cells via vanilloid receptors. Evidence for a
Held-Feindt, J., Dorner, L., Sahan, G., Mehdorn, H.M., Mentlein, R., 2006. Cannabi- protective role of cannabinoid receptors. J Biol Chem. 275 (41) 31938-45.
noid receptors in Human astroglial tumors. J. Neurochem. 98 (3), 886–893. Maccarrone, M., Cecconi, S., Rossi, G., Battista, N., Pauselli, R., Finazzi-Agrò, A., 2003.
Helliwell, R., Chamley, L., Blake-Palmer, K., Mitchell, M., Wu, J., Kearn, C., et al., Anandamide activity and degradation are regulated by early postnatal aging
2004. Characterization of the endocannabinoid system in early Human preg- and follicle-stimulating Hormone in mouse Sertoli cells. Endocrinology 144 (1),
nancy. J. Clin. Endocrinol. Metab. 89 (10), 5168–5174. 20–28.
Henshall, S., Afar, D., Hiller, J., Horvath, L., Quinn, D., Rasiah, K., et al., 2003. Survival Maione, S., Bisogno, T., De Novellis, V., Palazzo, E., Cristino, L., Valenti, M., et al.,
analysis of genome-wide gene expression profiles of prostate cancers identifies 2006. Elevation of endocannabinoid levels in the ventrolateral periaqueductal
New prognostic targets of disease relapse. Cancer Res. 63 (14), 4196–4203. grey through inhibition of fatty acid amide hydrolase affects descending noci-
Herkenham, M., Lynn, A.B., Johnson, M.R., Melvin, L.S., De Costa, B.R., Rice, K.C., ceptive pathways via both cannabinoid receptor type 1 and transient receptor
1991. Characterization and localization of cannabinoid receptors in rat brain: a potential vanilloid type-1 receptors. J. Pharmacol. Exp. Ther. 316 (3), 969–982.
quantitative in vitro autoradiographic study. J. Neurosci. 11 (2), 563–583. Maione, S., Costa, B., Piscitelli, F., Morera, E., De Chiaro, M., Comelli, F., et al., 2013.
Hoffman, A.F., Laaris, N., Kawamura, M., Masino, S.A., Lupica, C.R., 2010. Control of Piperazinyl carbamate fatty acid amide hydrolase inhibitors and transient re-
cannabinoid CB1 receptor function on glutamate axon terminals by en- ceptor potential channel modulators as "dual-target" analgesics. Pharmacol.
dogenous adenosine acting at A1 receptors. J. Neurosci. 30 (2), 545–555. Res. 76, 98–105.
Holland, M.L., Allen, J.D., Arnold, J.C., 2008. Interaction of plant cannabinoids with Malagarie-Cazenave, S., Olea-Herrero, N., Vara, D., Díaz-Laviada, I., 2009. Capsaicin,
the multidrug transporter ABCC1 (MRP1). Eur. J. Pharmacol. 591 (1–3), a component of red peppers, induces expression of androgen receptor via PI3K
128–131. and MAPK pathways in prostate LNCaP cells. FEBS Lett. 583 (1), 141–147.
Holland, M.L., Lau, D.T., Allen, J.D., Arnold, J.C., 2007. The multidrug transporter Malagarie-Cazenave, S., Olea-Herrero, N., Vara, D., Morell, C., Díaz-Laviada, I., 2011.
ABCG2 (BCRP) is inhibited by plant-derived cannabinoids. Br. J. Pharmacol. 152 The vanilloid capsaicin induces IL-6 secretion in prostate PC-3 Cancer cells.
(5), 815–824. Cytokine 54 (3), 330–337.
Hooijberg, J., Broxterman, H., Kool, M., Assaraf, Y., Peters, G., Noordhuis, P., et al., Malfitano, A.M., Ciaglia, E., Gangemi, G., Gazzerro, P., Laezza, C., Bifulco, M., 2011.
1999. Antifolate resistance mediated by the multidrug resistance proteins Update on the endocannabinoid system as an anticancer target. Expert Opin.
MRP1 and MRP2. Cancer Res. 59, 2532–2535. Ther. Targets 15 (3), 297–308.
Howlett, A.C., Barth, F., Bonner, T.I., Cabral, G., Casellas, P., Devane, W.A., et al., 2002. Mallat, A., Lotersztajn, S., 2008. Cannabinoid receptors as novel therapeutic targets
A comprehensive update on cannabinoid receptors and their biochemistry and for the management of non-alcoholic steatohepatitis. Diabetes Metab. 34 (6 Pt
pharmacology. international union of pharmacology. XXVII. Classification of 2), 680–684.
cannabinoid receptors. Pharmacol. Rev. 54 (2), 161–202. Martinez-Martinez, E., Gomez, I., Martin, P., Sanchez, A., Roman, L., Tejerina, E.,
Hurley, M.J., Mash, D.C., Jenner, P., 2003. Expression of cannabinoid CB1 receptor Bonilla, F., Merino, A.G., Herreros, A.G., Provencio, M., García, J.M., 2015. Can-
mRNA in basal ganglia of normal and parkinsonian Human brain. J. Neural nabinoids receptor type 2, CB2, expression correlates with Human colon Cancer
Transm. 110 (11), 1279–1288. progression and predicts patient survival. Oncoscience 2 (2), 131–141.
Ihenetu, K., Molleman, A., Parsons, M.E., Whelan, C.J., 2003. Inhibition of inter- Massi, P., Vaccani, A., Ceruti, S., Colombo, A., Abbracchio, M.P., Parolaro, D., 2004.
leukin-8 release in the Human colonic epithelial cell line HT-29 by cannabi- Antitumor effects of cannabidiol, a nonpsychoactive cannabinoid, on Human
noids. Eur. J. Pharmacol. 458 (1–2), 207–215. glioma cell lines. J. Pharmacol. Exp. Ther. 308 (3), 838–845.
Islam, T.C., Asplund, A.C., Lindvall, J.M., Nygren, L., Liden, J., Kimby, E., et al., 2003. Massi, P., Vaccani, A., Bianchessi, S., Costa, B., Macchi, P., Parolaro, D., 2006. The non-
High level of cannabinoid receptor 1, absence of regulator of G protein sig- psychoactive cannabidiol triggers caspase activation and oxidative stress in
nalling 13 and differential expression of cyclin D1 in mantle cell lymphoma. Human glioma cells. Cell Mol. Life Sci. 63 (17), 2057–2066.
Leukemia 17 (9), 1880–1890. Matsuda, L.A., Lolait, S.J., Brownstein, M.J., Young, A.C., Bonner, T.I., 1990. Structure
Iuvone, T., De Filippis, D., Di Spiezio Sardo, A., D'Amico, A., Simonetti, S., Sparice, S., of a cannabinoid receptor and functional expression of the cloned cDNA. Nature
et al., 2008. Selective CB2 up-regulation in women affected by endometrial 346 (6284), 561–564.
inflammation. J. Cell Mol. Med. 12 (2), 661–670. McAllister, S.D., Soroceanu, L., Desprez, P.Y., 2015. The anti-tumour activity of plant-
Izzo, A.A., Camilleri, M., 2009. Cannabinoids in intestinal inflammation and Cancer. derived non-psychoactive cannabinoids. J. Neuroimmune Pharmacol. 10,
Pharmacol. Res. 60 (2), 117–125. 255–267.
Jacobsson, S.O., Rongård, E., Stridh, M., Tiger, G., Fowler, C.J., 2000. Serum-depen- McKallip, R.J., Lombard, C., Fisher, M., Martin, B.R., Ryu, S., Grant, S., Nagarkatti, P.S.,
dent effects of tamoxifen and cannabinoids upon C6 glioma cell viability. Bio- Nagarkatti, M., 2002. Targeting CB2 cannabinoid receptors as a novel therapy to
chemPharmacol. 60 (12), 1807–1813. treat malignant lymphoblastic disease. Blood 100 (2) 627-34.
Jung, C.K., Kang, W.K., Park, J.M., Ahn, H.J., Kim, S.W., Taek, S.O.H., et al., 2013. Ex- McKallip, R.J., Nagarkatti, M., Nagarkatti, P.S., 2005. Delta-9-tetrahydrocannabinol
pression of the cannabinoid type I receptor and prognosis following surgery in enhances breast Cancer growth and metastasis by suppression of the antitumor
colorectal Cancer. Oncol. Lett. 5 (3), 870–876. immune response. J. Immunol. 174 (6), 3281–3289.
Karászi, E., Jakab, K., Homolya, L., Szakács, G., Holló, Z., Telek, B., et al., 2001. Calcein Mechoulam, R., Panikashvili, D., Shohami, E., 2002. Cannabinoids and brain injury:
assay for multidrug resistance reliably predicts therapy response and survival therapeutic implications. Trends Mol. Med. 8 (2), 58–61.
rate in acute myeloid leukaemia. Br. J. Haematol. 112, 308–314. Melck, D., Rueda, D., Galve-Roperh, I., De Petrocellis, L., Guzman, M., Di Marzo, V.,
Keppler, D., Leier, I., Jedlitschky, G., 1997. Transport of glutathione conjugates and 1999. Involvement of the cAMP/protein kinase a pathway and of mitogen-ac-
glucuronides by the multidrug resistance proteins MRP1 and MRP2. Biol. Chem. tivated protein kinase in the anti-proliferative effects of anandamide in Human
378 (8), 789–791. breast Cancer cells. FEBS Lett. 463 (3), 235–240.
Kim, J.A., Cho, K.B., Kim, M.R., Park, B.C., Kim, S.K., Lee, M.Y., et al., 2008. Decreased Melck, D., De Petrocellis, L., Orlando, P., Bisogno, T., Laezza, C., Bifulco, M., et al.,
production of vascular endothelial growth factor in adriamycin-resistant breast 2000. Suppression of nerve growth factor Trk receptors and prolactin receptors
Cancer cells. Cancer Lett. 268 (2), 225–232. by endocannabinoids leads to inhibition of Human breast and prostate Cancer
Kim, M.R., Choi, H.S., Yang, J.W., Park, B.C., Kim, J.A., Kang, K.W., 2009. Enhancement cell proliferation. Endocrinology 141 (1), 118–126.
F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14 13

Messalli, E.M., Grauso, F., Luise, R., Angelini, A., Rossiello, R., 2014. Cannabinoid 273–282.
receptor type 1 immunoreactivity and disease severity in Human epithelial Portenoy, R., Ganae-Motan, E., Allende, S., Yanagihara, R., Shaiova, L., Weinstein, S.,
ovarian tumors. Am. J. Obstet. Gynecol. 211 (3) 234.e231–234.e236. et al., 2012. Nabiximols for opioid-treated Cancer patients with poorly-con-
Mimeault, M., Pommery, N., Wattez, N., Bailly, C., Henichart, J.P., 2003. Anti-pro- trolled chronic pain: a randomized, placebo-controlled, graded-dose trial. J.
liferative and apoptotic effects of anandamide in Human prostatic Cancer cell Pain 13 (5), 438–449.
lines: implication of epidermal growth factor receptor down-regulation and Porter, A.C., Felder, C.C., 2001. The endocannabinoid nervous system: unique op-
ceramide production. Prostate 56 (1), 1–12. portunities for therapeutic intervention. Pharmacol. Ther. 90 (1), 45–60.
Molina-Holgado, E., Vela, J.M., Arevalo-Martin, A., Almazan, G., Molina-Holgado, F., Preet, A., Ganju, R.K., Groopman, J.E., 2008. Delta9-Tetrahydrocannabinol inhibits
Borrell, J., et al., 2002. Cannabinoids promote oligodendrocyte progenitor sur- epithelial growth factor-induced lung Cancer cell migration in vitro as well as
vival: involvement of cannabinoid receptors and phosphatidylinositol-3 kinase/ its growth and metastasis in vivo. Oncogene 27 (3), 339–346.
Akt signaling. J. Neurosci. 22 (22), 9742–9753. Preet, A., Qamri, Z., Nasser, M.W., Prasad, A., Shilo, K., Zou, X., et al., 2011. Canna-
Mon, M., Jansing, R., Doggett, S., Stein, J., Stein, G., 1978. Influence of delta9-tetra- binoid receptors, CB1 and CB2, as novel targets for inhibition of non-small cell
hydrocannabinol on cell proliferation and macromolecular biosynthesis in lung Cancer growth and metastasis. Cancer Prev. Res. 4 (1), 65–75.
Human cells. Biochem. Pharmacol. 27 (13), 1759–1765. Proto, M.C., Gazzerro, P., Di Croce, L., Santoro, A., Malfitano, A.M., Pisanti, S., et al.,
Morales, P., Blasco-Benito, S., Andradas, C., Gómez-Cañas, M., Flores, J.M., Goya, P., 2012. Interaction of endocannabinoid system and steroid hormones in the
Fernández-Ruiz, J., Sánchez, C., Jagerovic, N., 2015. Selective, nontoxic CB control of colon Cancer cell growth. J. Cell Physiol. 227 (1), 250–258.
(2) cannabinoid o-quinone with in vivo activity against triple-negative breast Qamri, Z., Preet, A., Nasser, M.W., Bass, C.E., Leone, G., Barsky, S.H., et al., 2009.
Cancer. J. Med. Chem. 58 (5), 2256–2264. Synthetic cannabinoid receptor agonists inhibit tumor growth and metastasis
Moreno, E., Andradas, C., Medrano, M., Caffarel, M.M., Perez-Gomez, E., Blasco- of breast Cancer. Mol. Cancer Ther. 8 (11), 3117–3129.
Benito, S., et al., 2014. Targeting CB2-GPR55 receptor heteromers modulates Qin, N., Neeper, M., Liu, Y., Hutchinson, T., Lubin, M., Flores, C., 2008. TRPV2 is ac-
Cancer cell signaling. J. Biol. Chem. 289 (32), 21960–21972. tivated by cannabidiol and mediates CGRP release in cultured rat dorsal root
Morioka, N., Abdin, J.M., Morita, K., Kitayama, T., Nakata, Y., Dohi, T., 2008. The ganglion neurons. J. Neurosci. 28 (24), 6231–6238.
regulation of glycine transporter GLYT1 is mainly mediated by protein kinase Ramer, R., Hinz, B., 2008. Inhibition of Cancer cell invasion by cannabinoids via
Calpha in C6 glioma cells. Neurochem. Int. 53 (6–8), 248–254. increased expression of tissue inhibitor of matrix metalloproteinases-1. J. Natl.
Mukhopadhyay, B., Schuebel, K., Mukhopadhyay, P., Cinar, R., Godlewski, G., Xiong, Cancer Inst. 100 (1), 59–69.
K., et al., 2015. Cannabinoid receptor 1 promotes hepatocellular carcinoma in- Ramer, R., Merkord, J., Rohde, H., Hinz, B., 2010. Cannabidiol inhibits Cancer cell
itiation and progression through multiple mechanisms. Hepatology 61 (5), invasion via upregulation of tissue inhibitor of matrix metalloproteinases-1.
1615–1626. Biochem. Pharmacol. 79 (7), 955–966.
Munro, S., Thomas, K.L., Abu-Shaar, M., 1993. Molecular characterization of a per- Ramer, R., Fischer, S., Haustein, M., Manda, K., Hinz, B., 2014. Cannabinoids inhibit
ipheral receptor for cannabinoids. Nature 365 (6441), 61–65. angiogenic capacities of endothelial cells via release of tissue inhibitor of matrix
Munson, A.E., Harris, L.S., Friedman, M.A., Dewey, W.L., Carchman, R.A., 1975. An- metalloproteinases-1 from lung Cancer cells. Biochem. Pharmacol. 91 (2),
tineoplastic activity of cannabinoids. J. Natl. Cancer Inst. 55 (3), 597–602. 202–216.
Nasser, M.W., Qamri, Z., Deol, Y.S., Smith, D., Shilo, K., Zou, X., et al., 2011. Crosstalk Ramer, R., Heinemann, K., Merkord, J., Rohde, H., Salamon, A., Linnebacher, M.,
between chemokine receptor CXCR4 and cannabinoid receptor CB2 in mod- et al., 2013. COX-2 and PPAR-gamma confer cannabidiol-induced apoptosis of
ulating breast Cancer growth and invasion. PLoS One 6 (9), e23901. Human lung Cancer cells. Mol. Cancer Ther. 12 (1), 69–82.
Norris, M., Bordow, S., Marshall, G., Haber, P., Cohn, S., Haber, M., 1996. Expression Richfield, E.K., Herkenham, M., 1994. Selective vulnerability in Huntington disease:
of the gene for multidrug-resistance-associated protein and outcome in pa- preferential loss of cannabinoid receptors in lateral globus pallidus. Ann.
tients with neuroblastoma. N. Engl. J. Med. 334, 231–238. Neurol. 36 (4), 577–584.
Notarnicola, M., Messa, C., Orlando, A., Bifulco, M., Laezza, C., Gazzerro, P., et al., Robson, P., 2011. Abuse potential and psychoactive effects of δ-9-tetra-
2008. Estrogenic induction of cannabinoid CB1 receptor in Human colon Cancer hydrocannabinol and cannabidiol oromucosal spray (Sativex), a New cannabi-
cell lines. Scand. J. Gastroenterol. 43 (1), 66–72. noid medicine. Expert Opin. Drug Saf. 10 (5), 675–685.
Nunez, E., Benito, C., Pazos, M.R., Barbachano, A., Fajardo, O., Gonzalez, S., et al., Romero, J., Berrendero, F., Garcia-Gil, L., De la Cruz, P., Ramos, J.A., Fernandez-Ruiz, J.
2004. Cannabinoid CB2 receptors are expressed by perivascular microglial cells J., 1998. Loss of cannabinoid receptor binding and messenger RNA levels and
in the Human brain: an immunohistochemical study. Synapse 53 (4), 208–213. cannabinoid agonist-stimulated [35S]guanylyl-5′O-(thio)-triphosphate binding
Olea-Herrero, N., Vara, D., Malagarie-Cazenave, S., Diaz-Laviada, I., 2009. Inhibition in the basal ganglia of aged rats. Neuroscience 84 (4), 1075–1083.
of Human tumour prostate PC-3 cell growth by cannabinoids R( þ )-Metha- Ross, R.A., 2003. Anandamide and vanilloid TRPV1 receptors. Br. J. Pharmacol. 140
nandamide and JWH-015: involvement of CB2. Br. J. Cancer 101 (6), 940–950. (5), 790–801.
Ortega, A., Rangel-López, E., Hidalgo-Miranda, A., Morales, A., Ruiz-García, E., Rossato, M., Ion Popa, F., Ferigo, M., Clari, G., Foresta, C., 2005. Human sperm ex-
Meneses-García, A., 2015. On the effects of CP 55-940 and other cannabinoid press cannabinoid receptor CB1, the activation of which inhibits motility, ac-
receptor agonists in C6 and U373 cell lines. Toxicol Vitr. 29 (7), 1941–1951. rosome reaction, and mitochondrial function. J. Clin. Endocrinol. Metab. 90 (2),
Pacher, P., Batkai, S., Kunos, G., 2006. The endocannabinoid system as an emerging 984–991.
target of pharmacotherapy. Pharmacol. Rev. 58 (3), 389–462. Rubovitch, V., Gafni, M., Sarne, Y., 2004. The involvement of VEGF receptors and
Paria, B., Song, H., Wang, X., Schmid, P., Krebsbach, R., Schmid, H., et al., 2001. MAPK in the cannabinoid potentiation of Ca2 þflux into N18TG2 neuro-
Dysregulated cannabinoid signaling disrupts uterine receptivity for embryo blastoma cells. Brain Res. Mol. 120 (2), 138–144.
implantation. J. Biol. Chem. 276 (23), 20523–20528. Ruiz, L., Miguel, A., Diaz-Laviada, I., 1999. Delta9-tetrahydrocannabinol induces
Park, B., Gibbons, H., Mitchell, M., Glass, M., 2003. Identification of the CB1 can- apoptosis in Human prostate PC-3 cells via a receptor-independent mechanism.
nabinoid receptor and fatty acid amide hydrolase (FAAH) in the Human pla- FEBS Lett. 458 (3), 400–404.
centa. Placenta 24 (10), 990–995. Ruiz-Llorente, L., Sanchez, M.G., Carmena, M.J., Prieto, J.C., Sanchez-Chapado, M.,
Patsos, H.A., Hicks, D.J., Dobson, R.R., Greenhough, A., Woodman, N., Lane, J.D., et al., Izquierdo, A., et al., 2003. Expression of functionally active cannabinoid re-
2005. The endogenous cannabinoid, anandamide, induces cell death in color- ceptor CB1 in the Human prostate gland. Prostate 54 (2), 95–102.
ectal carcinoma cells: a possible role for cyclooxygenase 2. Gut 54 (12), Sánchez, A., Sánchez, M., Malagarie-Cazenave, S., Olea, N., Díaz-Laviada, I., 2006.
1741–1750. Induction of apoptosis in prostate tumor PC-3 cells and inhibition of xenograft
Perez-Gomez, E., Andradas, C., Flores, J.M., Quintanilla, M., Paramio, J.M., Guzman, prostate tumor growth by the vanilloid capsaicin. Apoptosis 11 (1), 89–99.
M., et al., 2013. The orphan receptor GPR55 drives skin carcinogenesis and is Sailler, S., Schmitz, K., Jager, E., Ferreiros, N., Wicker, S., Zschiebsch, K., et al., 2014.
upregulated in Human squamous cell carcinomas. Oncogene 32 (20), Regulation of circulating endocannabinoids associated with Cancer and me-
2534–2542. tastases in mice and humans. Oncoscience 1 (4), 272–282.
Pertwee, R.G., 2006. The pharmacology of cannabinoid receptors and their ligands: Sánchez, C., De Ceballos, M.L., Gomez del Pulgar, T., Rueda, D., Corbacho, C., Velasco,
an overview. Int. J. Obes. 30 (Suppl 1), S13–S18. G., et al., 2001a. Inhibition of glioma growth in vivo by selective activation of the
Pertwee, R.G., 2009. Emerging strategies for exploiting cannabinoid receptor ago- CB(2) cannabinoid receptor. Cancer Res. 61 (15), 5784–5789.
nists as medicines. Br. J. Pharmacol. 156 (3), 397–411. Sánchez, C., Rueda, D., Segui, B., Galve-Roperh, I., Levade, T., Guzman, M., 2001b. The
Pertwee, R.G., Ross, R.A., Craib, S.J., Thomas, A., 2002. (  )-Cannabidiol Antagonizes CB(1) cannabinoid receptor of astrocytes is coupled to sphingomyelin Hydro-
cannabinoid receptor agonists and noradrenaline in the mouse vas deferens. lysis through the adaptor protein Fan. Mol. Pharmacol. 59 (5), 955–959.
Eur. J. Pharmacol. 456 (1–3), 99–106. Sánchez, M., Sanchez, A., Collado, B., Malagarie-Cazenave, S., Olea, N., Carmena, M.,
Pertwee, R.G., Howlett, A.C., Abood, M.E., Alexander, S.P., Di Marzo, V., Elphick, M.R., et al., 2005. Expression of the transient receptor potential vanilloid 1 (TRPV1) in
et al., 2010. International union of BASIC and clinical pharmacology. LXXIX. LNCaP and PC-3 prostate Cancer cells and in Human prostate tissue. Eur. J.
Cannabinoid receptors and their ligands: beyond CB1 and CB2. Pharmacol. Rev. Pharmacol. 515 (1–3), 20–27.
62 (4), 588–631. Sanchez, M.G., Sanchez, A.M., Ruiz-Llorente, L., Diaz-Laviada, I., 2003. Enhancement
Petersen, G., Moesgaard, B., Schmid, P.C., Schmid, H.H., Broholm, H., Kosteljanetz, of androgen receptor expression induced by (R)-methanandamide in prostate
M., et al., 2005. Endocannabinoid metabolism in Human glioblastomas and LNCaP cells. FEBS Lett. 555 (3), 561–566.
meningiomas compared to Human non-tumour brain tissue. J. Neurochem. 93 Santoro, A., Pisanti, S., Grimaldi, C., Izzo, A.A., Borrelli, F., Proto, M.C., et al., 2009.
(2), 299–309. Rimonabant inhibits Human colon Cancer cell growth and reduces the for-
Pineiro, R., Maffucci, T., Falasca, M., 2011. The putative cannabinoid receptor GPR55 mation of precancerous lesions in the mouse colon. Int. J. Cancer 125 (5),
defines a novel autocrine loop in Cancer cell proliferation. Oncogene 30 (2), 996–1003.
142–152. Sarfaraz, S., Afaq, F., Adhami, V.M., Mukhtar, H., 2005. Cannabinoid receptor as a
Pisanti, S., Picardi, P., D'Alessandro, A., Laezza, C., Bifulco, M., 2013. The en- novel target for the treatment of prostate Cancer. Cancer Res. 65 (5),
docannabinoid signaling system in Cancer. Trends Pharmacol. Sci. 34 (5), 1635–1641.
14 F.A. Javid et al. / European Journal of Pharmacology 775 (2016) 1–14

Sarfaraz, S., Afaq, F., Adhami, V.M., Malik, A., Mukhtar, H., 2006. Cannabinoid re- Van Sickle, M.D., Duncan, M., Kingsley, P.J., Mouihate, A., Urbani, P., Mackie, K., et al.,
ceptor agonist-induced apoptosis of Human prostate Cancer cells LNCaP pro- 2005. Identification and functional characterization of brainstem cannabinoid
ceeds through sustained activation of ERK1/2 leading to G1 cell cycle arrest. J. CB2 receptors. Science 310 (5746), 329–332.
Biol. Chem. 281 (51), 39480–39491. Vara, D., Salazar, M., Olea-Herrero, N., Guzman, M., Velasco, G., Diaz-Laviada, I.,
Sarfaraz, S., Adhami, V.M., Syed, D.N., Afaq, F., Mukhtar, H., 2008. Cannabinoids for 2011. Anti-tumoral action of cannabinoids on hepatocellular carcinoma: role of
Cancer treatment: progress and promise. Cancer Res. 68 (2), 339–342. AMPK-dependent activation of autophagy. Cell Death Differ. 18 (7), 1099–1111.
Sarnataro, D., Pisanti, S., Santoro, A., Gazzerro, P., Malfitano, A.M., Laezza, C., et al., Velasco, G., Sanchez, C., Guzman, M., 2012. Towards the use of cannabinoids as
2006. The cannabinoid CB1 receptor antagonist rimonabant (SR141716) inhibits antitumour agents. Nat. Rev. Cancer 12 (6), 436–444.
Human breast Cancer cell proliferation through a lipid raft-mediated me- Velasco, G., Hernandez-Tiedra, S., Davila, D., Lorente, M., 2015. The use of canna-
chanism. Mol. Pharmacol. 70 (4), 1298–1306. binoids as anti-cancer agents. Prog. Neuro-Psychopharmacol. Biol. Psychiatry
Schley, M., Stander, S., Kerner, J., Vajkoczy, P., Schupfer, G., Dusch, M., et al., 2009. 64, 259–266.
Predominant CB2 receptor expression in endothelial cells of glioblastoma in Velasco, L., Ruiz, L., Sanchez, M.G., Diaz-Laviada, I., 2001. Delta(9)-Tetra-
humans. Brain Res. Bull. 79 (5), 333–337. hydrocannabinol increases nerve growth factor production by prostate PC-3
Scott, K.A., Dalgleish, A.G., Liu, W.M., 2014. The combination of cannabidiol and cells. Involvement of CB1 cannabinoid receptor and Raf-1. Eur. J. Biochem. 268
Delta9-tetrahydrocannabinol enhances the anticancer effects of radiation in an (3), 531–535.
orthotopic murine glioma model. Mol. Cancer Ther. 13 (12), 2955–2967. Wang, D., Wang, H., Ning, W., Backlund, M.G., Dey, S.K., DuBois, R.N., 2008. Loss of
Shire, D., Calandra, B., Rinaldi-Carmona, M., Oustric, D., Pessegue, B., Bonnin-Ca- cannabinoid receptor 1 accelerates intestinal tumor growth. Cancer Res. 68
banne, O., et al., 1996. Molecular cloning, expression and function of the murine (15), 6468–6476.
CB2 peripheral cannabinoid receptor. Biochim. Biophys. Acta 1307 (2), 132–136. Wang, H., Guo, Y., Wang, D., Kingsley, P., Marnett, L., Das, S., et al., 2004. Aberrant
Shrivastava, A., Kuzontkoski, P.M., Groopman, J.E., Prasad, A., 2011. Cannabidiol cannabinoid signaling impairs oviductal transport of embryos. Nat. Med. 10
induces programmed cell death in breast Cancer cells by coordinating the (10), 1074–1080.
cross-talk between apoptosis and autophagy. Mol. Cancer Ther. 10, 1161–1172. Wasik, A.M., Nygren, L., Almestrand, S., Zong, F., Flygare, J., Wennerholm, S.B., et al.,
Skaper, S.D., Buriani, A., Dal Toso, R., Petrelli, L., Romanello, S., Facci, L., et al., 1996. 2014. Perturbations of the endocannabinoid system in mantle cell lymphoma:
The Aliamide palmitoylethanolamide and cannabinoids, but not anandamide, correlations to clinical and pathological features. Oncoscience 1 (8), 550–557.
are protective in a delayed postglutamate paradigm of excitotoxic death in Westlake, T.M., Howlett, A.C., Bonner, T.I., Matsuda, L.A., Herkenham, M., 1994.
cerebellar granule neurons. Proc. Natl. Acad. Sci. USA 93 (9), 3984–3989.
Cannabinoid receptor binding and messenger RNA expression in Human brain:
Sloan, E.K., Stanley, K.L., Anderson, R.L., 2004. Caveolin-1 inhibits breast Cancer
an in vitro receptor autoradiography and in situ hybridization histochemistry
growth and metastasis. Oncogene 23 (47), 7893–7897.
study of normal aged and Alzheimer brains. Neuroscience 63 (3), 637–652.
Smart, D., Jerman, J.C., 2000. Anandamide: an endogenous activator of the vanilloid
White, A.C., Munson, J.A., Munson, A.E., Carchman, R.A., 1976. Effects of delta9-
receptor. Trends Pharmacol. Sci. 21 (4), 134, Apr..
tetrahydrocannabinol in lewis lung adenocarcinoma cells in tissue culture. J.
Solinas, M., Massi, P., Cinquina, V., Valenti, M., Bolognini, D., Gariboldi, M., et al.,
Natl. Cancer Inst. 56 (3), 655–658.
2013. Cannabidiol, a non-psychoactive cannabinoid compound, inhibits pro-
Wijnholds, J., Scheffer, G., Van der Valk, M., Van der Valk, P., Beijnen, J., Scheper, R.,
liferation and invasion in U87-MG and T98G glioma cells through a multitarget
et al., 1998. Multidrug resistance protein 1 protects the oropharyngeal mucosal
effect. PLoS One 8 (10), e76918.
layer and the testicular tubules against drug-induced damage. J. Exp. Med. 188,
Stella, N., 2004. Cannabinoid signaling in glial cells. Glia 48 (4), 267–277.
797–808.
Sullivan, J.M., 2000. Cellular and molecular mechanisms underlying learning and
Wijnholds, J., DeLange, E., Scheffer, G., Van den Berg, D., Mol, C., Van der Valk, M.,
memory impairments produced by cannabinoids. Learn. Mem. 7 (3), 132–139.
et al., 2000. Multidrug resistance protein 1 protects the choroid plexus epi-
Sun, Y., Bennett, A., 2007. Cannabinoids: a new group of agonists of PPARs. PPAR
Res. 2007, 23513. thelium and contributes to the blood-cerebrospinal fluid barrier. J. Clin. Invest.
Thapa, D., Lee, J.S., Heo, S.W., Lee, Y.R., Kang, K.W., Kwak, M.K., et al., 2011. Novel 105 (3), 279–285.
hexahydrocannabinol analogs as potential anti-cancer agents inhibit cell pro- Williams, T.M., Medina, F., Badano, I., Hazan, R.B., Hutchinson, J., Muller, W.J., et al.,
liferation and tumor angiogenesis. Eur. J. Pharmacol. 650 (1), 64–71. 2004. Caveolin-1 gene disruption promotes mammary tumorigenesis and
Tomiyama, K., Funada, M., 2011. Cytotoxicity of synthetic cannabinoids found in dramatically enhances lung metastasis in vivo. role of Cav-1 in cell invasiveness
'spice’ products: the role of cannabinoid receptors and the caspase cascade in and matrix metalloproteinase (MMP-2/9) secretion. J. Biol. Chem. 279 (49),
the NG 108-15 cell line. Toxicol. Lett. 207, 12–17. 51630–51646.
Torres, S., Lorente, M., Rodriguez-Fornes, F., Hernandez-Tiedra, S., Salazar, M., Gar- Wright, K., Rooney, N., Feeney, M., Tate, J., Robertson, D., Welham, M., et al., 2005.
cia-Taboada, E., et al., 2011. A combined preclinical therapy of cannabinoids and Differential expression of cannabinoid receptors in the Human colon: canna-
temozolomide against glioma. Mol. Cancer Ther. 10 (1), 90–103. binoids promote epithelial wound healing. Gastroenterology 129 (2), 437–453.
Tsavaler, L., Shapero, M., Morkowski, S., Laus, R., 2001. Trp-p8, a novel prostate- Xu, X., Liu, Y., Huang, S., Liu, G., Xie, C., Zhou, J., et al., 2006. Overexpression of
specific gene, is up-regulated in prostate Cancer and other malignancies and cannabinoid receptors CB1 and CB2 correlates with improved prognosis of
shares high homology with transient receptor potential calcium channel pro- patients with hepatocellular carcinoma. Cancer Genet. Cytogenet. 171 (1),
teins. Cancer Res. 61 (9), 3760–3769. 31–38.
Tsou, K., Brown, S., Sanudo-Pena, M.C., Mackie, K., Walker, J.M., 1998. Im- Zhang, L., Barritt, G., 2004. Evidence that TRPM8 is an androgen-dependent
munohistochemical distribution of cannabinoid CB1 receptors in the rat Central Ca2 þ channel required for the survival of prostate Cancer cells. Cancer Res. 64
nervous system. Neuroscience 83 (2), 393–411. (22), 8365–8373.
Vaccani, A., Massi, P., Colombo, A., Rubino, T., Parolaro, D., 2005. Cannabidiol in- Zhu, L.X., Sharma, S., Stolina, M., Gardner, B., Roth, M.D., Tashkin, D.P., et al., 2000.
hibits Human glioma cell migration through a cannabinoid receptor-in- Delta-9-tetrahydrocannabinol inhibits antitumor immunity by a CB2 receptor-
dependent mechanism. Br. J. Pharmacol. 144 (8), 1032–1036. mediated, cytokine-dependent pathway. J. Immunol. 165 (1), 373–380.
Valverde, O., Karsak, M., Zimmer, A., 2005. Analysis of the endocannabinoid system Ziglioli, F., Frattini, A., Maestroni, U., Dinale, F., Ciufifeda, M., Cortellini, P., 2009.
by using CB1 cannabinoid receptor knockout mice. Handb. Exp. Pharmacol. 168, Vanilloid-mediated apoptosis in prostate Cancer cells through a TRPV-1 de-
117–145. pendent and a TRPV-1-independent mechanism. Acta Biomed. 80 (1), 13–20.

You might also like